溶血磷脂酸受体3和环氧合酶2及子宫内膜胞饮突与小鼠子宫内膜容受性的相关研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
胚胎植入是妊娠成功的关键因素之一,子宫内膜容受性的建立是胚胎成功植入的决定因素。因此对子宫内膜胚胎容受性的研究显得尤为重要。子宫内膜容受性是指子宫内膜在卵巢甾体激素调节下,子宫内膜形态、组织结构和分泌蛋白等发生一序列变化,使子宫内膜处于一种允许囊胚定位、黏附、侵入并使内膜间质发生改变从而导致胚胎着床的状态。这个特定的时期称为“着床窗期”。此时子宫内膜和胚胎相互作用,在非常短的着床窗期共同完成着床过程。在此前后,子宫内膜对胚胎处于不容受状态。在这一过程中,众多因素包括类固醇激素、生长因子、细胞因子信号以及黏附分子等,他们在不同的时间和空间环境控制下表达并发挥作用,共同构成了支持胚胎成功植入的分子基础。任何一个或几个因素表达水平出现异常,都有可能导致内膜环境发生改变,影响子宫内膜容受性,从而导致不孕。目前,子宫内膜容受性建立的确切机制尚不甚清楚,这对提高助孕技术水平和调节生育方法的发展是极大的障碍,进一步了解参与调节子宫内膜容受性的信号途径,将有助于我们更好地理解胚胎种植失败的原因,从而提高临床妊娠率。
     溶血磷脂酸(Lysophosphatidic acid LPA)是Mills等在卵巢癌腹水中发现的一种新的生长因子,为细胞膜脂类衍生物,是细胞间磷脂类信号分子。存在于机体的血清、眼房水和血浆等多种体液中,主要由活化的血小板、成纤维细胞和脂肪细胞以及癌细胞分泌,也可由某些炎症细胞、内皮细胞等产生。LPA主要通过与靶细胞上特异G蛋白偶联受体结合,发挥多种生物学功能:包括细胞的生存、增殖、分化、细胞骨架的重排、细胞间的相互作用以及肿瘤的形成和侵袭等。LPA受体主要有四种(Lysophosphatidic acid receptor LPAR)LPAR1-4,属内皮分化基因家族。LPAR1、LPAR2在机体内广泛存在,而LPAR3主要表达于卵巢、心脏、睾丸、前列腺等组织。最近研究发现,LPAR1、LPAR2主要参与小鼠神经系统的发育、颅面的形成,与神经系统疾病密切相关;LPAR3则主要与雌性生殖系统有关。敲除LPAR3基因的雌性小鼠,表现为胚胎种植延迟、分布空间异常、子宫内膜环氧合酶-2(Cyclooxygenase-2 COX-2)、前列腺素E2(prostaglandin E2 PGE2)、前列腺素I2(prostaglandin I2 PGI2)表达降低。COX-2是合成前列腺素的限速酶。对该基因缺失小鼠若给予外源性PGE2和PGI2类似物--前列环素(carbaprostacyclin cPGI),可有效改善LPAR3基因缺失所致的胚胎种植延迟,提示LPA可能通过LPAR3-COX2-PGS下游信号途径调节胚胎种植。目前对LPAR3研究较多的是其在肿瘤侵袭、转移中的作用。最新研究表明,卵巢癌高表达LPA及其受体LPAR2~3,可刺激肿瘤细胞的增殖、迁移、粘附、基质金属蛋白酶(matrix metalloproteinase,MMP)活化和血管生成因子的分泌,促进肿瘤细胞的侵袭转移。胚胎着床过程与肿瘤细胞的侵袭过程有一定的相似之处。目前对LPAR3在子宫内膜容受性的建立和胚胎着床方面的研究甚少。
     胞饮突是扫描电镜下所见的子宫内膜上皮细胞膜顶端出现的大而平滑的膜状突起。在啮齿类动物,它的出现与子宫内膜着床窗完全一致。在人类,它出现在月经中期LH峰的第6~8d,正是囊胚开始与子宫内膜粘附的时期。所以,胞饮突被认为是代表子宫内膜容受性的特异性形态学标记物。发育完全的胞饮突一般只存在24小时,代表了暂时性的细胞状态。目前有关胞饮突调节生殖作用的研究较多,但与LPAR3间关系的研究,国内外尚未见报道。
     控制性超促排卵(Controlled ovarian hyperstimulation COH)是决定体外受精-胚胎移植技术(In vitro fertilization-embryo transfer IVF–ET)成功与否的重要环节。在此过程中,为了获得多个同步发育的成熟卵子,常应用促性腺激素释放激素激动剂(Gonadotropin releasing hormone agonist GnRHa)和促排卵药物。目前临床上最常用GnRHa长方案超促排卵。大量研究显示,该方案能有效改善卵子质量,提高卵子回收率,受精率及优质胚胎率,但胚胎种植率和临床妊娠率仍较低。多数学者认为,这可能与GnRHa长方案超排卵后,干扰了内源性激素的平衡,影响了子宫内膜形态及分泌功能,使内膜发育与胚胎发育不同步,从而降低了子宫内膜容受性有关。但COH影响子宫内膜容受性的确切机制还不清楚。因此,探讨子宫内膜容受性降低的机制并寻找有效的拮抗方法,对提高IVF-ET的临床妊娠率将有着重要意义。那么,GnRHa长方案促排卵是否影响子宫内膜LPAR3及胞饮突的表达及发育?目前国内外未见报道。
     因此,研究LPAR3、COX-2在小鼠围着床期子宫内膜中的表达、作用及其调节机制以及与胞饮突的关系,可能会为探讨子宫内膜容受性的建立,揭示COH影响子宫内膜容受性的分子机制,提供理论依据。
     受人类临床取材的制约,本研究选用昆明小鼠作为研究对象,因小鼠和人类同属哺乳动物,着床过程类似。
     第一部分溶血磷脂酸受体3和环氧合酶-2在小鼠围着床期子宫内膜的表达及胞饮突发育的研究
     目的:探讨溶血磷脂酸受体3(Lysophosphatidic acid receptor 3 LPAR3) mRNA及其蛋白,环氧合酶(Cyclooxygenase-2 COX-2)在小鼠围着床期子宫内膜的表达以及胞饮突的发育变化。
     方法:采用半定量RT-PCR和Western blotting法以及免疫组织化学SP法,检测真孕组和假孕组小鼠LPAR3 mRNA及其蛋白在围着床期子宫内膜组织的表达规律。应用免疫组织化学SP法检测COX-2蛋白在子宫内膜的表达。扫描电子显微镜观察小鼠围着床期子宫内膜胞饮突的发育。
     结果:LPAR3 mRNA及其蛋白在动情期未孕、真孕、假孕组小鼠的子宫内膜均呈阳性表达。LPAR3蛋白主要分布于小鼠子宫内膜腔上皮、腺上皮和部分基质细胞的胞浆。在真孕组,LPAR3 mRNA及其蛋白性交后第3天(3 days post coitus 3d.p.c)表达开始明显上升,4 d.p.c达峰值,5 d.p.c、6 d.p.c骤然下降至基础水平。假孕组与相应同期真孕组的表达水平及变化规律一致( P > 0.05)。COX-2主要分布于小鼠子宫内膜腔上皮、腺上皮和基质细胞及血管内皮细胞的胞浆,在动情期未孕和真、假孕1~3 d.p.c不表达,4 d.p.c开始表达,并随妊娠天数增加,其表达逐渐向基质细胞延伸。基质细胞阳性染色增多、增强。假孕组和真孕组表达规律一致。胞饮突的发育模式:3 d.p.c为发育中的胞饮突,4 d.p.c出现完全发育的胞饮突,5 d.p.c为退化的胞饮突。假孕组和真孕组的发育模式一致。LPAR3 mRNA及其蛋白在子宫内膜上皮的表达峰值与完全发育的胞饮突的出现,在时间和部位上存在一致性。
     结论: LPAR3、COX-2和胞饮突可能参与了胚胎的着床及子宫内膜容受性的建立过程。LPAR3和子宫内膜的超微结构-胞饮突,表达时序一致,可作为判断子宫内膜具有容受性的分子参考指标。LPAR3和胞饮突的变化与宫腔内是否有胚胎存在无显著相关性,说明主要受母体信号的调节。COX-2可能参与胚胎植入过程。
     第二部分雌孕激素对小鼠子宫内膜及Ishikawa细胞LPAR3表达的影响
     目的:探讨雌、孕激素和孕激素拮抗剂米非司酮(RU486)对去势雌性小鼠在体子宫内膜及体外培养的子宫内膜腺癌细胞株Ishikawa细胞LPAR3 mRNA及其蛋白表达的影响。
     方法:建立去势雌性昆明小鼠动物模型,随机分为雌激素组,孕激素组,雌孕激素联合组以及孕激素+RU486组,应用半定量RT-PCR、Western blotting法,检测子宫内膜LPAR3 mRNA及其蛋白的表达。体外培养高分化子宫内膜腺癌细胞Ishikawa细胞,分别加入不同浓度17-β雌二醇(10-9M, 10-8M, 10-7M)、孕激素(10-7M, 10-6M, 10-5M),雌+孕激素(10-8M+10-6M)。采用免疫细胞化学方法检测给药刺激48 h后LPAR3蛋白的表达。
     结果:去势雌性小鼠子宫内膜表达LPAR3 mRNA;孕激素单独作用,可显著提高小鼠子宫内膜LPAR3 mRNA的表达,呈时间依赖性,但无明显量效关系;雌激素单独作用,对LPAR3 mRNA表达无显著影响。雌、孕激素联合应用,雌激素降低孕激素对子宫内膜LPAR3 mRNA的表达上调作用。RU486减弱孕激素的表达上调作用,但无明显量效关系。同组LPAR3蛋白的表达与基因表达趋势一致。各浓度雌激素、孕激素及雌孕激素联合应用,对体外培养的Ishikawa细胞LPAR3蛋白的表达均无明显影响( P > 0.05)。
     结论:在体动物实验:孕激素单独作用,显著提高小鼠子宫内膜LPAR3 mRNA及其蛋白的表达,呈时间依赖性,无明显量效关系;雌激素单独作用对小鼠子宫内膜LPAR3表达无明显影响;雌、孕激素联合作用,雌激素降低孕激素对LPAR3表达上调作用;RU486减弱孕激素的表达上调作用。雌、孕激素对Ishikawa细胞,LPAR3蛋白的表达无显著影响。
     第三部分GnRHa长方案促排卵对围着床期小鼠子宫内膜LPAR3的表达及胞饮突发育的影响
     目的:研究GnRHa/HMG/HCG控制性超促排卵(Controlled ovarian hyperstimulation COH)对小鼠子宫内膜LPAR3 mRNA及其蛋白表达的影响以及胞饮突发育的变化。探讨COH长方案促排卵影响子宫内膜容受性的可能分子机制,为临床改善促排卵方案提供理论依据。
     方法:将雌性健康未孕昆明系小鼠随机分为COH组和自然受孕组(natural conception NC),用半定量RT-PCR、Western blotting方法,检测3 d.p.c~6d.p.c子宫内膜LPAR3 mRNA及其蛋白的表达,同时应用扫描电子显微镜观察子宫内膜胞饮突发育情况。全自动免疫测定仪检测小鼠血清雌、孕激素水平。
     结果:COH组:3d.p.c LPAR3 mRNA及其蛋白的表达出现峰值,而NC组,孕4d.p.c达峰值。其余相应各时间点两组间LPAR3 mRNA及其蛋白的表达量无显著差异。COH组:3d.p.c胞饮突发育不同步,大小不一,有的为发育完全的胞饮突,有的为正在发育的胞饮突,表面不光滑,有微绒毛,多为发育不完全的胞饮突。胞饮突数量较对照组明显减少,呈局灶性分布。4d.p.c胞饮突发育不同步,大小不一,表面开始皱缩。5d.p.c子宫内膜大部分为被覆微绒毛的上皮细胞,可见少量退化的胞饮突。NC组:3d.p.c部分子宫内膜细胞表面开始突起,膜状突起表面覆盖微绒毛,绒毛短,稀疏,为发育中胞饮突;孕4d.p.c均匀分布着大量大小一致,发育同步,边界清楚的蘑菇状胞饮突,表面光滑。5d.p.c胞饮突表面皱缩,为退化的胞饮突。血清雌、孕激素水平:血清雌、孕激素在两组均随妊娠天数增加而升高;COH组各时间点雌孕激素浓度均高于相应同期NC组,两者比较有显著差异。( P < 0.05)
     结论:COH影响小鼠内源性激素的平衡,使雌、孕激素异常升高。可能通过改变与子宫内膜容受性相关的分子和超微结构的时序变化影响子宫内膜容受性,从而影响着床。
Embryo implantation is one of the vital factors for successful pregnancy and the establishment of endometrial receptiblity is the determinative factor for a well embryo implantation. As a result, the reseach on endometrial embryo receptiblity is very important. The endometrial receptiblity is a special condition of endometrium, during this period a series of changes in morphosis and secreted protein within the endometrium occurre under the control of ovarial steriod homone. As a consequence the allowance is available of apposition, adhesion, invasion and embryo implantation by the alteration in endometrial mesenchymal. This special period aslo be called“the window of implantation”. The implantation procedure can accomplish in this short peroid by the reaction between endometrium and embryo. On the contrary, the implantation is impossible before or behind this peroid. It can be drawed from researchs on endometrial receptiblity that many factors play important roles in the implantation procedure, which include steroid hormone, growth factor, cell factor signal, adhesion molecule etc. those factors expression and action in different time and under the control of different space enviromental form the molecular basis of embryo implantation. The expression level of any factors is abnormal will contribute to change the enviroment of endometrium and the endometrial receptivity and lead to infertility. The knowledge about the mechanism of implantation has not completely understood. It becomes the great handicape to improve the assisted reproductive technology level. To explore the pathway about the signal that participate the moderation procedure of the endometrial receptiblity will benefit us to get information about the failure of embryo implantation and increase the pregnancy rate accordingly.
     Lysophosphatidic acid (LPA)-a new growth factor, has been founded by Mills in abdominal fluids resulted from ovrian cancer, is a kind of derivation of cell membran lipid functioning as an intercell phospholipid signal. LPA, secreted by actived blood platelet, cancer cell, fibroblast and adipocyte or produced by some inflammatory cell and endothelial cell, exists in blood serum, ocular humor and blood plasma. LPA mainly exhibit multiple biological functions by connecting to the special G-protein couple receptor in the target cell that invovlved in many cellular processes including cellular survice, proliferation, differentiation, cytoskeletal reorganization, interaction with cell and tumour formation and cancer cell invasion and so on in many physiological and pathological process.There are at least four receptors for LPA, LPAR1-4. LPAR1 and LPAR2 are expressed in almost all tissues,but LPAR3 shows a limited expression pattern in the ovary, heart, testes and prostate. The lately research showed, LPAR1 and LPAR2 are participate in the development of neurology and the formation of craniofacial. They are closely correlated with neurologic disease. LPAR3 in particular is important in female reproductive system: The female mouse whose PLAR3 gene have been knocked out appeared a delayed embryo implantation, abnormal space distribution, decreased expression of COX-2 gene. COX-2 is a rate-limiting enzyme of prostaglandin synthesis. The fact that giving exogenous PGE2 and cPGI can compensate the delayed embryo implantation in LPAR3 gene absent showing the closed relationship between LPAR3, COX-2 and embryo implantation. Nowadays, the majority of researchs on LPA and its receptor were focus on the function in tumor invasion and migration. The latest study indicated that high level expression of PLA and its receptor LPAR2, LPAR3 can promote cancer cellular proliferation, migration, adhension and induce the activation of matrix metalloproteinase (MMP) and the secretion of angiogenesis factor in objects with ovarian cancer. The process of implantation was similar, in some sense, to the process of tumor cell invasion and migration. In the generation domain, there is lack of information about the function of LPAR3 on establishing endometrial receptiblity and embryo implantation until now.
     Pinopodes is a smooth bigger size prominence formed by endometrial epithelium. The occurrence of completely developed pinopodes during implantation peroid is regarded as the morphological characteristic of endometrial receptiblity which existed only for 24 hours and match the endometrial receptivity period. In rodent, pinopodes accordance to the window of implantation. In huaman, mature pinopodes are seen only during a brief window following the midcycle luteinizing hormone (LH) surge between LH+6 and LH+8. It is the period that blastocyst begin to attach to endometrium. As an ultramicrostructure marker of implantation window openning, about pinopodes there have many correlated studies in its function on reproduction. But no literature can be found at home and abroad about the relationship with LPAR3.
     Controlled ovarian hyperstimulation (COH) can get many ovum by using ovulation stimulants, is an important element in determination of whether the IVF-ET can success or not. Long protocol controlled ovarian hyperstimulation is the most commonly used protocol by Gonadotrophin releasing hormone agonist. Researchs demonstrated that this plan can improve the quality of ovum and got many ovum but compared with the high retrieval rate, the clinical pregnancy rate still in a low level.This may be because that the GnRHa long time superovulation program put a negative effect in the balance of internal hormone which affected the morphous and the secrete function of endometrium and resulted in asynchronization between the developtment of the endometrium and embryo. Decrease the endometrial receptiblity. Now the mechanism about the relationship between COH and decreasing in endometrial receptiblity is still unclear. So to evaluate the mechanism of decreasing in endometrial receptiblity and find an effective method to solve the problem plays a significent role in increasing the pregnancy rate by IVF-ET. Now it is absent in the document about the temporal and spatial interrelation with GnRHa long protocol controlled ovarian hyperstimulation on the expression of LPAR3 and pinopodes in mouse uterine endometrium during periimplantation periode.
     In view of that, to study the expression of LPAR3 and COX-2 in uterine endometrium and the development of pinopodes during the periimplantation of mouse and the relationship of coexpression will provide clue of theory for the building endometrial receptivity and reveal the maybe molecule mechanism of endometrial receptivity.
     The specimens of humen are limited, kunming mice and human are all mammals, the procedure of embryo implantation is similar, so we select kunming mouse as our study objectives.
     The partΙ: The expression of LPAR3 and COX-2 in uterine endometrium during the period of periimplantation of mouse embryos and the development of pinopodes
     Object: To evaluate the dynamic change of lysophosphatidic acid receptor 3 mRNA, protein and distribution. The expression and location of COX-2 in mouse uterine endometrium during the period of periimplantation; To evaluate the pinopodes development in window of implantation.
     Method: The expression of LPAR3 mRNA and protein in uterine endometrium tissues during periimplantation of pregnant and pseudopregnant mice were detected by using semiquantitative of PT-PCR, Western blotting and immunohistochemistry methods. The COX-2 expression and location was detected by immunohistochemistry SP methods. Using scanning electronmicroscope to observe the development of pinopodes during periimplantation peroid.
     Result: LPAR3 mRNA and protein were positive in endometrium of unpregnancy, pregnancy and pseudopregnancy mouse. LPAR3 protein mainly distributed in luminal and glandular epithelium and part of stromal cells of mouse uterus. Around the embryo implantation procedure in mouse, the level of LPAR3 mRNA and protein began to increase on 3 days post coitus (3d.p.c), peak around 4d.p.c and returned suddenly to basal level on 5d.p.c and 6d.p.c. The temporal profile of LPAR3 mRNA in the uterus of pseudopregnancy mouse was similar to those observed in normal pregnant mouse. There was no significant different between the two groups ( P > 0.05). The tendency of LPAR3 protein in pregnant and pseudopregnancy was regularity to LPAR3 mRNA. The COX-2 mainly existed in luminal, glandular epithelium vascular endothelial cell and part of stromal cell, didn't express in normal and before 3d.p.c, but began to express on 4d.p.c and extend to stromal cell along with the progression of pregnancy. The expression of COX-2 increased and deepened positive staining in stromal cells. The mode expression of COX-2 in pregnancy mouse was similar to that of pseudopregnancy. The mode of pinopodes: developing pinopodes appeared on 3d.p.c; mature pinopodes on 4d.p.c and degenerated pinopodes on 5d.p.c.which was coincidence with the expression peak of LPAR3 in uterine endometrial. The pattern was similar in pregnancy and pseudopregnancy.
     Conclusion: LPAR3, COX-2 and pinopodes may participate in the building of endometrial receptiblity. The concordance of LPAR3 and pinopodes was in the temporal and spatial and could be as a marker in judgement of endometrial receptibity. There was no relationship between the expression of LPAR3, the development of pinopodes and whether or not the presentation of embryo in uterus.That showed they were mainly regulated by matermal signal. COX-2 possiblely participated in the procedure of embryo implantation.
     The partΠ: The regulatory effect of estradiol progesterone on the expression of LPAR3 in uterine endometrium tissue and Ishikawa cell lines
     Object: To investigate the regulatory effect of estradiol, progesteronge and progensterone antagonist RU486 on the expression of LPAR3 gene and protein in uterine endometrium and Ishikawa cell lines.
     Method: Ovariectomized kunming mice were randomly divided into four groups: estradiol group, progesterone group, combined with estradiol and progesterone group, progesterone and RU486 group.The expression of LPAR3 mRNA and protein in uterine endometrium were detected by using semiquantitative of PT-PCR, Western blotting methods. The highly differentiated endometril adenocarcinoma cells(Ishikawa cell line) were cultured in vitro with 17-beta estradiol (E2)(10-9M, 10-8M, 10-7M), progesterone (10-7M, 10-6M, 10-5M), E2 combined with progesterone (10-8M+10-6M), for 48 hours respectively and the expression of LPAR3 protein was detected by immunocytochemistrical methods.
     Result: There was a positive LPAR3 mRNA expression in ovariectomized Kunming mouse endometrium tissue; Progesterone alone could improve the expression of LPAR3 mRNA in mouse endometrium markedly and showed a time dependent; Estradiol alone could not put significantly effect on LPAR3 mRNA expression; Estradiol combined with progesterone could counteracted the up-regulation effect of progesterone; RU486 could decrease the up-regulation effect of progesterone. The tendency expression of LPAR3 protein has a positive correlation with the mRNA. There was no significant effect on the expression of LPAR3 protein in Ishikawa cell lines by variety concentration of estradiol, progesterone, estradiol combined with progesterone.
     Conclusion: In vivo animal experiment, progesterone alone could increase the expression of LPAR3, but estradiol alone had no effect on it. Estradiol combined with progesterone could counteracted the up-regulation effect of progesterone; RU486 could decrease the up-regulation effect of progesterone. There was no significant effect on the expression of LPAR3 protein in Ishikawa cell lines by estradiol and progesterone.
     The partШ: The effect of long protocol controlled ovarian hyperstimulation by GnRHa on the expression of LPAR3 and the development of pinopodes in mouse uterine endometrium during periimplantation period
     Object: To investigate the influence of controlled ovarian hyperstimulation by GnRHa/HMG/HCG long protocol treatment to the expression of LPAR3 mRNA and protein and the development of pinopodes; To evaluate the maybe molecular mechanism of adverse effect on endometrial receptivity by COH which may provide theory for selecting an ideal programe of ovarian hyperstimulation in clinical.
     Method: Healthy femal unpregnant Kunming mice were randomly divided into COH group and natural conception group(NC).The expession of LPAR3 mRNA and protein in uterine endometrium tissue was detected by PT-PCR and Western blotting during the periimplantation period. The development of pinopodes on 3d.p.c~6d.p.c by using scanning electron microscope. The estrogen and progesterone level in mouse blood serum was detected by Abbott Laboratories A4sym automatic immune radiomete.
     Result: The peak expression of LPAR3 mRNA and protein on 3d.p.c in COH group, which was similar to natural conception group on 4d.p.c. There was no significant difference in other days. In the COH group, on 3d.p.c, the development of pinopodes was out-of-step, which characteristic of variation in size and in alignment. Some were mature pinopodes, most were developing ones, which had an uneven surface and microvilli. The number of pinopodes in COH group was less than in natural conception group and the style of expression was focal. On 4d.p.c, the development of pinopodes were derangement and the size was vary and were out of sync. On 5d.p.c, there were few degenerated pinopodes and most endometrium was covered with microvilli cells. In natural conception group: on 4d.p.c, there were a lots of coincidence pinopodes, the development was synchron, homogeneous distribution, equal size, smooth surface and looked like mushroom which progectured the surface of cells. On 5d.p.c , the surface of pinopodes were introcession and were degenerated pinopodes. The concentration of estrogen and progesterone in blood serum ascensus obviously in both groups with the progress of pregnancy, but it was higher in COH group than in natural conception group at each time. There was significantly different between two groups.
     Conclusion: COH impacted the balance of endogenous hormone in mouse which resulting in an abnormal higher level of estrogen and progesteron. COH altered the endometrial receptibility through changing the relative factors for implantation and the change of ultrastructural in the temporal and spatial.
引文
1 Carson DD, Bagchi I, Dey SK et al. Embryo implantation. Dev Biol, 2000,223(2):217~237
    2 MoolenaarWH. Lysophospholipids in the limelight: autotaxin takes center stage. J Cell Bio,2002,158(2):197~199
    3 Contos J, Ishii I, Chun J,et al. Lysophosphatidic acid receptors. Mol Pharmacol , 2000,58(6),1188~1196
    4 Moolenaar WH. Bioactive lysophospholipids and their G proteincoupled receptors. Experimental Cell Research,1999,253(1):230~238
    5 Gardell SE, Dubin AE, Chun J, et al. Emerging medicinal roles for lysophospholipid signaling. Trends in Mol Med,2006,12(2):65~75
    6 Li C, Dandridge KS, Di A, et al. Lysophosphatidic acid inhibits cholera toxin-induced secretory diarrhea through CFTR-dependent protein interactions. Journal of Experimental Medicine,2005,202(7):975~986
    7 Ye X, Hama K, Contos J, et al. LPA3-mediated lysophosphatidic acid signalling in embryo implantation and spacing. Nature,2005,435(7038): 104~108
    8 Hu YL, Tee M K, Goetzl EJ, et al. Lysophosphatidic acid induction ofvascular endot helial growth factor expression in human ovarian cancer cells. J Natl Cancer Inst, 2001,93(10):762~768
    9 Mills GB,Moolenaar WH. The emerging role of lysophosphatidic acid in cancer. Nature, 2003,3(8):582~591
    10 Smith WL, Dewitt DL, Garavito RM. Cyclooxygenases: structural, cellular and molecular biology. Annu Rev Biochem,2000,69:145~182
    11 Hama K, Aoki J, Bandoh K, et al. Lysophosphatidic receptor, LPA3, is positively and negatively regulated by progesterone and estrogen in the mouse uterus. Life Sci,2006,79(18):1736~1740
    12 Atarzyna Kami′nska, Marta Wasielak, Iwona Bogacka et al. Quantitative expression of lysophosphatidic acid receptor 3 gene in porcine endometrium during the periimplantation period and estrous cycle. Prostaglandins & Other Lipid Mediators,2008,85(1-2):26~32
    13 Breyer RM, Kennedy CR, Zhang Y, et al. Structure function analyses of eicosanoid receptors: physiology and therapeutic implications. Ann N Y Acad Sci,2000,905:221~231
    14 Chakraborty I, Das SK, Wang J, et al. Developmental expression of the cyclo-oxygenase-1 and cyclooxygenase-2 genes in the peri-implantation mouse uterus and their differential regulation by the blastocyst and ovarian steroids. J Mol Endocrinol,1996,16(2):107~122
    15 Kim JJ, Wang J, Bambra C, et al. Expression of cyclooxygenase-1 and -2 in the baboon endometrium during the menstrual cycle and pregnancy. Endocrinology, 1999,140(6):2672~2678
    16 Marions L, Danielsson KG. Expression of cyclooxygenase in human endometrium during the implantation period. Mol Hum Reprod, 1999,5(10):961~965
    17 Lim H, Paria BC, Das SK, et al. Multiple female reproductive failures in cyclooxygenase 2-deficient mice. Cell,1997,91:197~208
    18 Lim H, Gupta RA, Ma WG, et al. Cyclo-oxygenase-2-derived prostacyclin mediates embryo implantation in the mouse via PPARdelta. Genes Dev, 1999,13(12):1561~1574
    19 Nikas G, Psychoyos A. Uterine pinopodes in peri-implantation human endometrium. Clinical relevance Ann N Y Acad Sci,1997,816:129~142
    20 Murphy CR. Understanding the apical surfacemarkers of receptvity: pinopods oruterodomes? Hum Reprod,2000,15(12):2451~2454
    21 Nikas G, Aghajianova L. Endometrial pinopodes: some more understanding on human implantation? Reprod Biomed Online,2002, 4[Suppl3]:18~23
    22 NikasG, Makrigiannakis A. Endometrial pinopodes and uterine receptivity. Annals of the New York Academy of Sciences,2003,997:120~123
    23 Stavreus-Evers A, Masironil B, Landgren BM, et al. Immunohistochemical localization of glutaredoxin and thioredoxin in human endometrium: a possible associationwith pinopodes. Mol Hum Reprod,2002,8(6):546~551
    24 Nardo LG, Nikas G, Makrigiannakis A, et al. Synchronous expression of pinopodes and alpha v beta 3 and alpha 4 beta integrins in the endometrial surface epithelium of normally menstruating women during the implantation window. J Reprod Med,2003,48(5):355~361
    25 Fazleabas AT, Straskova Z. Endometrial function: cell specific changes in the uterine environment. Mol Cell Endocrinol,2002,186(2):143~147
    26 Kabir-Salmani M, Shiokawa S, Akimoto Y, et al. Tissue transglutaminase at embryo-maternal interface. J Clin Endocrinol Metab,2005,90(8): 4694~4702
    27 Stavreus-Evers A, Koraen L, Zhang P, et al. Distribution of cyclooxygenase-1, cyclooxygenase-2, and cytosolic phospholipase A2 in the luteal phase human endometrium and ovary. Fertil Steril,2005,83(1): 156~162
    28 Gabardi S, Cerio J. Future immunosuppressive agents in solid-organ transplantation. Prog Transplant,2004,14(2):148~156
    1 Klentzeris LD. The role of endometrium in implantation. Hum Reprod , 1997,12[Suppl 11 ]:170~175
    2 Dey SK, Lim H, Das SK, et al. Molecular cues to implantation. Endocrinology Review,2004,25(3):341~373
    3 Tigyi G. Physiological responses to lysophosphatidic acid and related glycerol-phospholipids. Prostaglandins,2001,64(1-4):47~62
    4 Fukushima N, Chun J. The LPA receptors. Prostaglandins, 2001,64(1-4):47~62
    5 Ye X, Hama K, Contos J. et al. LPA3-mediated lysophosphatidic acid signalling in embryo implantation and spacing. Nature,2005,435 (7038):104~108
    6 Kotaro Hama, Junken Aoki, Koji Bandoh, et al. Lysophosphatidic receptor LPA3 is positively and negatively regulated by progesterone and estrogen in the mouse uterus. Life Sci,2006,79(18):1736~1740
    7 Nishida M. The Ishikawa cells from birth to the present. Hum Cell,2002,15(3):104~117
    8 Somkuti SG, Yuan L, Fritz MA, et al. Epidermal growth factor and sex steroids dynamically regulate a marker of endometrial receptivity in ishikawa cells. J Clin Endocrinol Metab,1997,82(7):2192~2197
    9 Hata H, Kuramoto H. Immunocytochemical determination of estrogen and progesterone receptors in human endometrial adenocarcinoma cells (Ishikawa cells). J Steroid Biochem Mol Biol,1992,42(2):201~210
    10 Lessey BA, Ilesanmi AO, Castelbaum AJ, et al. Characterization of the functional progesterone receptor in an endometrial adenocarcinoma cell line (Ishikawa): progesterone-induced expression of the alpha1 integrin. J Steroid Biochem Mol Biol,1996,59(1),31~39
    11 McCormack JT, Greenwald GS. Progesterone and oestradiol-17beta concentrations in the peripheral plasma during pregnancy in the mouse. J Endocrinol,1974,62(1),101~107
    12 Carson D, Bagchi I, Dey S, et al. Embryo implantation. Deve Biol, 2000, 223(2):217~237
    13 Bagchi IC , Li Q , Cheon YP. Role of steroid hormone regulated genes in implantation. Ann N Y Acad Sci,2001,943:68~76
    14 de Ziegler D, Fanchin R, de Moustier B, et al. The hormonal control of endometrial receptivity: estrogen (E2) and progesterone. J Reprod Immunol,1998,39(1-2):149~166
    15 Lessey BA. Two Pathways of Progesterone Action in the Human Endometrium: Implications for Implantation and Cont raception. Steroids,2003,68(10-13):809~815
    16 Hata H, Kuramoto H. Immunocytochemical determination of estrogen and progesterone receptors in human endometrial adenocarcinoma cells (Ishikawa cells). J Steriod Biochem Mol Biol,1992,42(2):201~210
    17 Somkuti SG, Yuan L, Fritz MA, et al. Epidermal growth factor and sex steroids dynamically regulate a marker of endometrial receptivity in Ishikawa cells. J Clin Endocrinol Metab,1997,82(7):2192~2197
    18 Lessey BA, Gui Y, Apparao KB, et al. Regulated expression ofheparin-binding EGF-like growth factor (HB-EGF) in the human endometrium: a potential paracrine role during implantation. Mol Reprod Dev,2002,62(4):446~455
    1 Lopata A. Implantation of the human embryo. Hum Reprod,1996,11:175~184
    2 Serafini P, Rocha AM, Osorio CT et al. Endometrial leukemia inhibitory factor as a predictor of pregnancy after in vitro fertilization. International Journal of Gynecology and Obstetrics,2008,102:23~27
    3 Creus M, Ordi J, Fabregues F,et al. The effect of different hormone therapies on integrin expression and pinopode formation in the human endometrium:a controlled study. Hum Reprod,2003,18:683~693
    4 Shi XQ ,ed. Modern laboratory animal science of medicine. Beijing : People′s Military Medical Press,2000
    5 Nyboe AA , Gianaroli L, Nygren KG. Assisted reproductive technology inEurope , 2000. Results generated from European registers by ESHRE. Hum Reprod,2004,19:490~503
    6 Devroey P, Bourgain C, Macklon NS, et al. Rep roductive biology and IVF: ovarian stimulation and endometrial recep tivity. Trends Endocrinol Metab,2004,15:84 ~ 90
    7 Mirkin S, Nikas G, Hsiu JG, et al. Gene expression profiles and end structural functional features of the priImplantation endometrium in natural and gonadotropin simulated cycles. J Clin Endocrinol Metab,2004,89:5742~5752
    8 Hadi FH, Chantler E, Anderson E, et al. Ovulat ion induction and endometrial steroid receptors. Hum Reprod,1994,9:2405~2 410
    9 Tavaniotou A, Smitz J, Bourgain C, et al. Ovulation induction disrupts luteal phase function. Ann N Y Acad Sci,2001,943:55~63
    10 Dey SK, Lim H, Das SK, et al. Molecular cues to implantation.Endocrine. Review, 2004,25:341~373
    11 Ma WG, Song H, Das SK,et al. Estrogen is a critical determinant that specifies the suration of the window of uterine teceptivety for implantation.Proc Natl Acad Sci USA,2003,100:2963~2968
    12 Tavaniotou A,Albano C,Smitz J,et al. Impact of ovarian stimulation on corpus luteum function and embryonic implantation. J Reprod Immunol,2002,55:123~130
    13 Bourgain C, Ubaldi F, Tavaniotou A, et al. Endometrial hormone receptors and proliferation index in the periovulatory phase of stimulated embryo transfer cycles in comparison with natrual cycles and relation to clinical pregnagnancy outcome.Fertil Steril,2002,78:237~244
    14 Curits Hewitt S, Goulding EH, Eddy EM et al. Studies using the estrogen receptor a knockout uterus demonstrate that implantation but not decidulization associated signaling is estrogen dependent. Biol Reprod,2002,67:1268~1277
    15 Stavreus-Evers A, Nikas G, Sahlin L, et al. Formation of pinopodes in human endometrium is associated with the concentrations ofprogesterone and p rogesterone receptors. Fertil Steril, 2001,76: 782~791
    16 Makker A, Tandon I, Goel MM et al. Effect of ormeloxifene, a selective estrogen receptor modulator, on biomakers of endometrial receptivity and pinopode development and its relation to fertility and infertility in Indian subjects. Fertil Steril,2008,Epub ahead of print
    17 Martel D, Monier MN, Roche D et al. Hormonal depengdent of pinopode formationat the uterine luminal surface. Hum Reprod,1991,6:579~603.
    18 Bourgain C. Endometrial biopsy in the evaluation of endometrial receptivity. J Gynecol Obstet Biol Reprol ( Paris), 2004,33:S13 ~17
    19 Ruan HC, Zhu XM, Luo Q, et al. Ovarian stimulation with GnRH agonist, but not GnRH antagonist, partially restores the expression of endometrial integrin beta3 and leukaemia-inhibitory factor and improves uterine receptivity in mice. Hum Reprod,2006,21:2521~2529
    20 Macklon NS, Fauser BC. Impact of ovarisn hyperstimulation on the luteal phase. J Reprod Fertil Suppl,2000,55:101~108
    21 Check JH, Choe JK, Katsoff D, et al. Controlled ovarian hyperstimulation adversely affects implantation following in vitro fertilizationembryo transfer. J Assist Reprod Genet,1999,16:416~420
    22 Ma WG, Song H, Das SK,et al. Estrogen is a critical determinant that specifies the duration of the window of uterine receptivity for implantation. Proc Natl Acad USA,2003,100:2963~2968
    23 Thomas K,Thomason AJ,Wood SJ,et al. Endometrial integrin expression in women undergoing IVF and ICSI:a comparison of the two groups and fertile. HumReprod,2003,18:364~369
    24 Ertzeid G, Storeng R. The impact of ovarian stimulation on implantation and fetal development in mice. Hum Reprod,2001,16:221~225
    25 Levi AJ, Drews MR, Bergh PA, et al. Controlled ovarian stimulation does not adversely affect endometrial receptivity in in vitro fertilization cycles.Fertil Steril,2001,76:670~674
    1 Santos WL, Rossi JA, Boggs SD, et al. The molecular pharmacology of lysophosphatide signaling. Ann N Y Acad Sci,2000,905:232~241
    2 Eichholtz T, Jalink K, Fahrenfort I, et al. The bioactive phospholipidlysophosphatidic acid is released from activated platelets. Biochem J,1993,291:677~680
    3 Dennis EA. Diversity of group types, regulation and function of phospholipase A2. J Biol Chem,1994,269:13057~13060
    4 Aoki J. Mechanisms of lysophosphatidic acid production. Semin Cell Dev Biol,2004,15:477~489
    5 Sugiura T, Nakane S, Kishimoto S, et al. Lysophosphatidic acid, a growth factor-like lipid, in the saliva. J Lipid Res,2002,43:2049~2055
    6 Hama K, Bandoh K, Kakehi Y, et al. Lysophosphatidic acid (LPA) receptors are activated differentially by biological fluids: possible role of LPA-binding proteins in activation of LPA receptors. FEBS Lett, 2002,523:187~192
    7 Tokumura A, Miyake M, Nishioka Y, et al. Production of lysophosphatidic acids by lysophospholipase D in human follicular fluids of in vitro fertilization patients. Biol Reprod,1999,61:195~199
    8 Tokumura A, Kanaya Y, Miyake M, et al. Increased production of bioactive lysophosphatidic acid by serum lysophospholipase D in human pregnancy. Biol Reprod,2002,67:1386~1392
    9 Fukushima N, Weiner JA, Chun J. Lysophosphatidic acid (LPA) is a novel extracellular regulator of cortical neuroblast morphology. Dev Biol,2000,228:6~18
    10 Pages C, Daviaud D, An S, et al. Endothelial differentiation gene-2 receptor is involved in lysophosphatidic acid-dependent control of 3T3F442A preadipocyte proliferation and spreading. J Biol Chem,2001,276:11599~11605
    11 Pages C, Simon MF, Valet P, et al. Lysophosphatidic acid synthesis and release. Prostaglandins Other Lipid Mediat,2001,64:1~10
    12 Weiner JA, Fukushima N, Contos JJ, et al. Regulation of Schwann cell morphology and adhesion by receptor-mediated lysophosphatidic acid signaling. J Neurosci ,2001,21:7069~7078
    13 Kitzing TM, Sahadevan AS, Brandt DT et al. Lysophosphatidic acid-stimulated interleukin-6 and -8 synthesis through lpa1 receptors on osteoblasts.Arch Oral Biol,2008,53:207~213
    14 Mauco G, Chap H, Simon MF, et al. Phosphatidic and lysophosphatidic acid production in phospholipase c-and thrombintreated platelets. Possible involvement of a platelet lipase. Biochimie,1978,60:653~661
    15 Shen Z, Belinson J, Morton RE, et al. Phorbol 12-myristate 13-acetate stimulates lysophosphatidic acid secretion from ovarian and cervical cancer cells but not from breast or leukemia cells. Gynecol Oncol,1998,71:364~368
    16 Eder AM, Sasagawa T, Mao M, et al. Constitutive and lysophosphatidic acid (lpa)-induced lpa production: role of phospholipased and phospholipase A2. Clin Cancer Res,2000,6:2482~2491
    17 le Balle F, Simon MF, Meijer S, et al. Membrane sidedness of biosynthetic pathways involved in the production of lysophosphatidic acid. Adv Enzyme Regul,1999,39:275~284
    18 Tokumura A, Majima E, Kariya Y, et al. Identification of human plasma lysophospholipase d, a lysophosphatidic acid-producing enzyme as autotaxin a multifunctional phosphodiesterase. J Biol Chem,2002,277:39436~39442
    19 Umezu-Goto M, Kishi Y, Taira A, et al. Autotaxin has lysophospholipase d activity leading to tumor cell growth and motility by lysophosphatidic acidproduction. J Cell Biol,2002,158:227~233
    20 Tanyi JL, Hasegawa Y, Lapushin R, et al. Role of decreased levels of lipid phosphate phosphatase-1in accumulation of lysophosphatidic acid in ovarian cancer. Clin Cancer Res,2003,10(Part 1):3534~3545
    21 Tanyi JL, Morris AJ, Wolf JK, et al. The human lipid phosphate phosphatase-3 decreases the growth,survival, and tumorigenesis of ovarian cancer cells: validation of the lysophosphatidic acid signaling cascade as a target for therapy in ovarian cancer. Cancer Res,2003,163:1073~1082
    22 Tigyi G. Physiological responses to lysophosphatidic acid and related glycerol-phospholipids. Prostaglandins,2001,64:47~62
    23 Fukushima N, Chun J. The LPA receptors. Prostaglandins,2001,64:21~32
    24 Meyer zu Heringdorf D, Jakobs KH. Lysophospholipid receptors: signalling, pharmacology and regulation by lysophospholipid metabolism. Biochim Biophys Acta,2007,1768:923~940
    25 Noguchi K, Ishii S, Shimizu T. Identification of p2y9/GPR23 as a novel G protein-coupled receptor for lysophosphatidic acid, structurally distant from the Edg family. J Biol Chem,2003,278:25600~25606
    26 Lee CW,Rivera R, Gardell S, et al. GPR92as anewG12/13- and Gq-coupled lysophosphatidic acid receptor that increases cAMP, LPA5. J Biol Chem,2006,281:23589~23597
    27 van Corven EJ, Groenink A, Jalink K, et al. Lysophosphatidate-induced cell proliferation: identification and dissection of signaling pathways mediated by G proteins. Cell,1989,59:45~54
    28 Contos JJ, Ishii I, Chun J. Lysophosphatidic acid receptors. Molecular Pharmacology,2000,58:1188~1196
    29 Gardell SE, Dubin AE, Chun J. Emerging medicinal roles for lysophospholipid signaling. Trends in Molecular Medicine,2006,12: 65~75
    30 Hu YL, Tee MK, Goetzl EJ, et al. Lysophosphatidic acid induction of vascular endothelial growth factor expression in human ovarian cancer cells. J Natl Cancer Inst,2001,93:762~768
    31 So J, Navari J, Wang FQ et al. Lysophosphatidic acid enhances epithelialovarian carcinoma invasion through the increased expression of interleukin-8. Gynecol Oncol,2004,95:314~322
    32 Chou CH, Wei LH, Kuo ML, et al. Up-regulation of interleukin-6 in human ovarian cancer cell via a Gi/PI3K–Akt/NF-_B pathway by lysophosphatidic acid, an ovarian cancer-activating factor. Carcinogenesis,2005,26:45~52
    33 Noguchi K, Herr D, Chun J. Lysophosphatidic acid and its receptors. Curr Opin Pharmacol,2009,9:15~23
    34 Fukushima N, Ishii I, Contos JJ, et al. Lysophospholipid receptors. Annu Rev Pharmacol Toxicol, 2001,58:507~534
    35 Moolenaar WH. Bioactive lysophospholipids and their g proteincoupled receptors. Exp Cell Res,1999,1:230~238
    36 Senqupta S, Wang Z, Tipps R et al. Biology of LPA in health and disease. Seminars in Cell & Developmental Biology,2004,15:503~512
    37 Waters CM, Saatian B, Moughal NA, et al. Integrin signalling regulates the nuclear localization and function of the lysophosphatidic acid receptor-1 (LPA1) in mammalian cells. Biochem,2006,398:55~62
    38 Zhao C, Fernandes MJ, Prestwich GD, et al. Regulation of lysophosphatidic acid receptor expression and function in human synoviocytes: implications for rheumatoid arthritis? Mol Pharmacol,2008,73:587~600
    39 Tokumura A. Metabolic pathways and physiological and pathological significances of lysolipid phosphate mediators. J Cell Biochem, 2004,92:869~881
    40 Xu YJ, Rathi SS, Chapman DC, et al. Mechanisms of lysophosphatidic acid-inducedDNAsynthesis in vascular smooth muscle cells. J Cardiovasc Pharmacol,2003,41:381~387
    41 Gobeil F Jr, Bernier SG, Vazquez-Tello A, et al. Modulation of pro-inflammatory gene expression by nuclear lysophosphatidic acid receptor type-1. J Biol Chem,2003,278:38875~38883
    42 Hu YL, Albanese C, Pestell RG, et al. Dual mechanisms forlysophosphatidic acid stimulation of human ovarian carcinoma cells.Cancer Ins,2003,95:733~740
    43 Kunikata K, Yamano S, Tokumura A, et al. Effect of lysophosphatidic acid on the ovum transport in mouse oviducts. Life Sci, 1999,65:833~840
    44 Kobayashi T, Yamano S, Murayama S, et al. Effect of lysophosphatidic acid on the preimplantation development of mouse embryos. FEBS Lett, 1994,351:38~40
    45 Jalink K, Hordijk PL, Moolenaar W,et al. Growth factor-like effects of lysophosphatidic acid, a novel lipid mediator. Biochimica et Biophysica Acta, 1994,1198:185~196
    46 Tokumura A. A family of phospholipid autacoids: occurrence metabolism and bioactions. Progress in Lipid Research,1995,34:151~184
    47 Tokumura A, Miyake M, Nishioka Y, et al. Production of lysophosphatidic acids by lysophospholipase D in human follicular fluids of in vitro fertilization patients. Biology of Reproduction,1999,61:195~199
    48 Hinokio K, Yamano S, Nakagawa K, et al. Lysophosphatidic acid stimulates nuclear and cytoplasmic maturation of golden hamster immature oocytes in vitro via cumulus cells. Life Sciences,2002,70:759~767
    49 Liu Z, Armant DR. Lysophosphatidic acid regulates murine blastocyst development by transactivation of receptors for heparin-binding EGF-like growth factor.Experimental Cell Research,2004,296:317~326
    50 Durieux ME, Salafranca MN, Lynch KR, et al. Lysophosphatidic acid induces a pertussis toxin-sensitive ca(2+)-activated cl-current in xenopus laevis oocytes. Am J Physiol,1992,263:C896~900
    51 Thomson FJ, Moyes C, Scott PH, et al. Lysophosphatidic acid stimulates glucose transport in xenopus oocytes via a phosphatidylinositol 3triphosphate-binding protein RhoA in the process of implantation. J Clin Endocrinol Metab,2000,85:4742~4749
    54 Tamada H, Higashiyama C, Takano H, et al. The effects of heparin-binding epidermal growth factor-like growth factor on preimplantation-embryo development and implantation in the rat. Life Sci, 1999,64:1967~1973
    55 Klonisch T, Wolf P, Hombach-Klonisch S, et al. Epidermal growth factor-like ligands and erbB genes in the peri-implantation rabbit uterus and blastocyst. Biol Reprod,2001,64:1835~1844
    56 Wang X,Wang H, Matsumoto H, et al. Dual source and target of heparin-binding EGF-like growth factor during the onset of implantation in the hamster. Development,2002,129:4125~4134
    57 Kanda Y, Mizuno K, Kuroki Y, et al. Thrombin-induced p38 mitogen-activated protein kinase activation is mediated by epidermal growth factor receptor transactivation pathway. British Journal of Pharmacology,2001,132:1657~1664
    58 Parast MM, Aeder S, Sutherland AE, et al. Trophoblast giant-cell differentiation involves changes in cytoskeleton and cell motility. Dev Biol, 2001,230:43~60
    59 Sah VP, Seasholtz TM, Sagi SA, et al. The role of Rho in G protein-coupled receptor signal transduction. Annu Rev Pharmacol Toxicol, 2000,40:459 ~489
    60 MoolenaarWH. Development of our current understanding of bioactive lysophospholipids. Ann NY Acad Sci,2000,905:1~10
    61 He D, Natarajan V, Stern R, et al. Lysophosphatidic acid-induced transactivation of epidermal growth factor receptor regulates cyclo-oxygenase-2 expression and prostaglandin E(2) release via C/EBP_ in human bronchial epithelial cells. Biochem,2008,412:153~162
    62 Oyesanya RA, Lee ZP, Wu J, et al. Transcriptional and post-transcriptional mechanisms for lysophosphatidic acid-induced cyclooxygenase-2 expression in ovarian cancer cells. FASEB,2008,22:2639~2651
    63 Nicola C, Chirpac A, Lala PK, et al. Roles of Rho guanosine 5_-triphosphatase A, Rho kinases, and extracellular signal regulated kinase (1/2) in prostaglandin E2-mediated migration of first-trimester human extravillous trophoblast. Endocrinology,2008,149:1243~1251
    64 Biondi C, Ferretti ME, Pavan B,et al. Prostaglandin E2 inhibits proliferation and migration of HTR-8/SVneo cells, a human trophoblast-derived cell line. Placenta,2006,27:592~601
    65 Chen SU, Lee H, Chang DY, et al. Lysophosphatidic Acid Mediates Interleukin-8 Expression in Human Endometrial Stromal Cells through Its Receptor and Nuclear Factor-κB-Dependent Pathway: A Possible Role in Angiogenesis of Endometrium and Placenta.Endocrinology, 2008,149:5888~5896
    66 Yang AH, Ishii I, Chun J. In vivo roles of lysophospholipid receptors revealed by gene targeting studies in mice. Biochim Biophys Acta, 2002,1582:197~203
    67 Contos JJ, Fukushima N, Weiner JA,et al. Requirement for the lpA1 lysophosphatidic acid receptor gene in normal suckling behavior. Proc Natl Acad Sci USA,2000,97:13384~13389
    68 Contos JJ, Ishii I, Fukushima N, et al. Characterization of lpa(2) (Edg4) and lpa(1)/lpa(2) (Edg2/ Edg4) lysophosphatidic acid receptor knockout mice: signaling deficits without obvious phenotypic abnormality attributable to lpa(2). Mol Cell Biol,2002,22:6921~6929
    69 Ye X, Hama K, Contos J , et al. LPA3-mediated lysophosphatidic acid signalling in embryo implantation and spacing. Nature, 2005,435:104~108
    70 Symowicz J, Adley BP, Woo M, et al. Cyclooxygenase-2 functions as a downstream mediator of lysophosphatidic acid to promote aggressive behavior in ovarian carcinoma cells. Cancer Res,2005,65:2234~2242
    71 Reiser C O, Lanz T, Hofmann F, et al. Lysophosphatidic acid-mediated signal-transduction pathways involved in the induction of the early-response genes prostaglandin G/H synthase-2 and Egr-1: a criticalrole for the mitogen-activated protein kinase p38 and for Rho proteins. Biochem,1998,330:1107~1114
    72 Lim H, Gupta RA, Ma WG, et al. Cyclo-oxygenase-2-derived prostacyclin mediates embryo implantation in the mouse via PPARdelta. Genes Dev, 1999,13:1561~1574
    73 Mohamed OA, Jonnaert M, Labelle-Dumais C, et al. Uterine Wnt/beta catenin signaling is required for implantation. Proc Natl Acad Sci USA, 2005,102:8579~8584
    74 Gschwind A, Hart S, Fischer OM, et al.TACE cleavage of proamphiregulin regulates GPCR-induced proliferation and motility of cancer cells. EMBO J,2003,22:2411~2421
    75 Symowicz J, Adley BP, Woo MM, et al. Cyclooxygenase-2 functions as a downstream mediator of lysophosphatidic acid to promote aggressive behavior in ovarian carcinoma cells. Cancer Res,2005,65:2234~2242
    76 Atarzyna Kami′nska , Marta Wasielak , Iwona Bogacka et al. Quantitative expression of lysophosphatidic acid receptor 3 gene in porcine endometrium during the periimplantation period and estrous cycle. Prostaglandins & Other Lipid Mediators,2008,85:26~32
    77 Heewon S, Mingoo K, Yohan C,et al. Analysis of Lysophosphatidic Acid (LPA) Receptor and LPA-Induced Endometrial Prostaglandin Endoperoxide Synthase 2 Expression in the Porcine Uterus. Endocrinology, 2008,149:6166~6175
    78 Gabardi S, Cerio J. Future immunosuppressive agents in solid-organ transplantation. Prog Transplant,2004,14:148~156
    1 Bentin Ley U,Sjogren A,Nilsson L,et al. Presence of uterine pinopodes at the embryo-endometrial interface during humanimplantation in vitro. Hum Reprod,1999,14:515~520
    2 Bentin Ley U. Relevance of endometrial pinopodes for human blastocyst implantation. Hum Reprod,2000,15[Suppl 6]:67~73
    3 Enders AC, Nelson DM. Pinocytotic activity of the uterus of the rat. Am J Anat,1973,138:277~299
    4 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). C R Acad Sci Hebd Seances Acad Sci D, 1971,272:2723~2725
    5 Nikas G, Drakakis P, Loutradis D, et al. Uterine pinopodes as markers of the‘nidation window’in cycling women receiving exogenous oestradiol and progesterone. Hum Reprod,1995,10:1208~1213
    6 Psychoyos A, Nikas G. Uterine pinopodes as markers of uterine receptivity. Assist Reprod Rev,1994,4:26~41
    7 Nikas G. Endometrial receptivity: changes in cell-surface morphology. Semin Reprod Med,2000,18:229~235
    8 Nikas G, Develioglu OH, Toner JP, et al. Endometrial pinopodes indicate a shift in the window of receptivity in IVF cycles. Hum Reprod, 1999,14:787~792
    9 Bentin-Ley U, Sjogren A, Nilsson L, et al. Presence of uterine pinopodes at the embryo–endometrial interface during human implantation in vitro. Hum Reprod,1999,14:515~20
    10 Nikas G, Makrigiannakis A, Hovatta O, et al. Surface morphology of the human endometrium. Basic and clinical aspects. Ann N Y Acad Sci, 2000,900:316 ~324
    11 Aghajanova L, Stavreus-Evers A, Nikas Y, et al. Coexpression ofpinopodes and leukemia inhibitory factor, as well as its receptor, in human endometrium. Fertil Steril,2003,79(Suppl 1):808~814
    12 Pantos K, Nikas G, Makrakis E, et al. Clinical value of endometrial pinopodes detection in artificial donation cycles. Reprod Biomed Online, 2004,9:86~90
    13 Quinn C, Ryan E, Claessens EA et al. The presence of pinopodes in the human endometrium does not delineate the implantation window. Fertil Steril,2007,87:1015~1021
    14 Nikas G, Drakakis P, Loutradis D, et al. Uterine pinopodes as markers of the“nidation window”in cycling women receiving exogenous oestradiol and progesterone. Hum Reprod,1995,10:1208~1213
    15 Stavreus-Evers A, Nikas G, Sahlin L, et al. Formation of pinopodes in human endometrium is associated with the concentrations of progesterone and progesterone receptors. Fertil Steril,2001,76:782~791
    16 Stavreus-Evers A, Mandelin E, Koistinen R, et al. Glycodelin is present in pinopodes of receptive-phase human endometrium and is associated with down-regulation of progesterone receptor B. Fertil Steril, 2006,85:1803~1811
    17 Ordi J, Creus M, Casamitjana R, et al. Endometrial pinopode and alphavbeta3 integrin expression is not impaired in infertile patients with endometriosis. J Assist Reprod Genet,2003,20:465~473
    18 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 cyclingwomen with proven fertility. Fertil Steril,2003,79:970~974
    19 Oborna I, Novotny R, Brezinova J, et al. Changes in the development of uterine pinopodes in steroid hormone supplemented cycles. Physiol Res, 2004,53:423~429
    20 Petersen A, Bentin-Ley U, Ravn V, et al. The antiprogesterone Org 31710 inhibits human blastocyst endometrial interactions in vitro. Fertil Steril, 2005,83(Suppl 1):1255~1263
    21 Lessey BA, Damjanovich L, Coutifaris C, et al. Integrin adhesion molecules in the human endometrium. Correlation with the normal and abnormal menstrual cycle. J Clin Invest,1992,90:188~195
    22 Lessey BA, Ilesanmi AO, Lessey MA, et al. Luminal and glandular endometrial epithelium express integrins differentially throughout the menstrual cycle: Implications for implantation, contraception, and infertility. Am J Reprod Immunol,1996,35:195~204
    23 Lessey BA, Castelbaum AJ, Buck CA, et al. Further characterization of endometrial integrins during the menstrual cycle and in pregnancy. Fertil Steril,1994,62:497~506
    24 Wilcox AJ, Baird DD, Wenberg CR. Time of implantation of the conceptus and loss of pregnancy. N Engl J Med,1999,340:1796~1799
    25 Savaris RF, Pedrini JL, Flores R, et al. Expression of alpha 1 and beta 3 integrins subunits in the endometrium of patients with tubal phimosis or hydrosalpinx.Fertil Steril,2006,85:188~192
    26 Thomas K, Thomson A, Wood S, et al. Endometrial integrin expression in women undergoing in vitro fertilization and the association with subsequent treatment outcome. Fertil Steril,2003,80:502~507
    27 Nardo LG, Bartoloni G, Di Mercurio S, et al. Expression of the alpha(v)beta3 and alpha4beta1 integrins throughout the putative window of implantation in a cohort of healthy fertile women. Acta Obstet Gynecol Scand,2002,81:753~758
    28 Bildirici I, Bukulmez O, Ensari A, et al. A prospective evaluation of the effect of salpingectomy on endometrial receptivity in cases of women with communicating hydrosalpinges. Hum Reprod,2001,16:2422~2426
    29 von Wolff M, Strowitzki T, Becker V, et al. Endometrial osteopontin, a ligand of beta3-integrin, is maximally expressed around the time of the implantation window. Fertil Steril,2001,76:775~781
    30 Garcia P, Nieto A, Sanchez MA, et al. Expression of alpha4, alpha5 and beta3 integrin subunits, fibronectin and vitronectin in goat-periimplantation. Anim Reprod Sci,2004,80:91~100
    31 Lin H, Wang X, Liu G, et al. Expression of alphaV and beta3 integrin subunits during implantation in pig. Mol Reprod Dev,2007,74:1379~1385
    32 Lessey BA, Castelbaum AJ. Integrins in the endometrium of women with endometriosis. Br J Obstet Gynaecol,1995,102:347~348
    33 Gonzalez RR, Palomino A, Boric A, et al. A quantitative evaluation of alpha1, alpha4, alphaV and beta3 endometrial integrins of fertile and unexplained infertile women during the menstrual cycle. A flow cytometric appraisal. Hum Reprod,1999,14:2485~2492
    34 Illera MJ, Lorenzo PL, Gui YT, et al. A role for alphavbeta3 integrin during implantation in the rabbit model.Biol Reprod,2003,68:766~771
    35 Illera MJ, Cullinan E, Gui Y, et al. Blockade of the alpha(v)beta(3) integrin adversely affects implantation in the mouse. Biol Reprod, 2000,62:1285~1290
    36 Srinivasan KR, Blesson CS, Fatima I, et al. Expression of aVb3 integrin in rat endometrial epithelial cells and its functional role during implantation. Gen Comp Endocrinol,2009,160:124~133
    37 Nardo LG, Bartoloni G, Di Mercurio S, et al. Expression of alpha(v) beta3 and alpha4beta1 integrins throughout the putative window of implantation in a cohort of healthy fertile women. Acta Obstet Gynecol Scand,2002, 81: 753~758
    38 Kimmins S, Lim HC, Parent J, et al. The effects of estrogen and progesterone on prostaglandins and integrin beta 3 (beta3) subunit expression in primary cultures of bovine endometrial cells. Domest Anim Endocrinol,2003,25:141~154
    39 Lessey BA, Gui Y, Apparao KB, et al. Regulated expression of heparin-binding EGF-like growth factor (HB-EGF) in the human endometrium: a potential paracrine role during implantation. Mol Reprod Dev,2002,62:446~455
    40 Daftary GS, Troy PJ, Bagot CN, et al. Direct regulation of beta3 integrin subunit gene expression by HOXA10 in endometrial cells Mol Endocrinol, 2002,16:571~579
    41 Chen QJ, Sun XX, Li L,et al. Effects of ovarian stimulation on endometrial integrin beta3 and leukemia inhititory factor expression in the peri-implantation phase.Fertil Steril,2008,89(5 suppl):1357~1363
    42 Bulletti C, Flamigni C, de Ziegler D. Implantation markers and endometriosis. Reprod Biomed Online,2005,11:464~468
    43 Kabir-Salmani M, Nikzad H, Shiokawa S, et al. Secretory role for human uterodomes (pinopods): secretion of LIF. Mol Hum Reprod, 2005,11:553~559
    44 Smith S, Pfeifer SM, Collins JA. Diagnosis and management of female infertility. JAMA,2003,290:1767~1770
    45 Stewart CL, Kaspar P, Brunet LJ, et al. Blastocyst implantation depends on maternal expression of leukaemia inhibitory factor. Nature 1992,359:76–79
    46 Hambartsoumian E. Endometrial leukemia inhibitory factor (LIF) as a possible cause of unexplained infertility and multiple failures of implantation. Am J Reprod Immunol,1998,39:137~143
    47 Laird SM, Tuckerman EM, Dalton CF,et al. The production of leukaemia inhibitory factor by human endometrium: presence in uterine flushings and production by cells in culture. Hum Reprod,1997,12:569~574
    48 Aqhajanova L, Altmae S,Bjuresten K, et al. Disturbances in the LIF pathway in the endometrium among women with unexplained infertility. Fertil Steril,2008,4(Epub ahead of print)
    49 Paulo S, AndréM. Rocha C,et al. Endometrial leukemia inhibitory factor as a predictor of pregnancy after in vitro fertilization. Int J Gynecol Obstet,2008,102:23~27
    50 Brinsden PR, Ndukwe G, Engrand P, et al. Does recombinant human leukemia inhibitory factor improve implantation in women with recurrent failure of assisted reproduction treatment? In 19th Annual Meeting of the European Society for Human Reproduction and Embryology. Madrid, Spain,2003,pp.18
    51 Sawai K, Matsuzaki N, Okada T, et al. Human deciual cell biosynthesis ofleukemia inhibitory factor regulation by decidual cutokines and steroid hormones.Biol Reprod,1997,56:1274~1280
    52 Quinn CE, Detmar J, Casper RF. Pinopodes are present in Lif null and Hoxa10 null mice.Fertil Steril,2007,88:1021~1028
    53 Bagot CN, Kliman HJ, Taylor HS. Maternal Hoxa10 is required for pinopod formation in the development ofmouse uterine receptivity to embryo implantation. Dev Dyn,2001,222:538~544
    54 Bagot CN, Troy PJ, Taylor HS, et al. Alteration of maternal HOXA10 expression by in vivo gene transfection affects implantation. Gene Ther, 2000,7:1378~1384
    55 Kim JJ, Taylor HS, Lu Z, et al. Altered expression of HOXA10 in endometriosis: potential role in decidualization. Mol Hum Reprod, 2007,13:323~332
    56 Dickey RP,Olar TT, Taylor SN, et al. Relationship of Endometrial thickness and Patternt to fencundity in ovalation Induction cycles:Effect of clomiphene citrate alone and with Human menopausal gonadotropin.Fertil Steril,1993,59:756~760
    57 Bourgain C, Devroey P. The endometrium in stimulated cycles for IVF.Hum Reprod Update,2003,9:515~22
    58 Zaidi J, Campbell S,Tan SL. Endometrial thickness morphology vascular penetration and velocimetry in predicting implantion in an in vitro fetilization program.Ultrasound Obstet Gynecol,1995,6:191~198
    59 Tsai HD, Chang CC, Hsieh YY, et al. Artificial insemination. Role of endometrial thickness and pattern of vascular impedance of the spiral and uterine arteries and of the dominant follicle. J Reprod Med, 2000,45:195~200
    60 Wakeman S, Benny P.Is it possible to predict a fertile cycle?Uteroovarian blood flow patameters in conception versus nonconception cycles.Fertil Steril,2008,17:249~258
    61 Wu HM, Chiang CH, Huang HY,et al. Detection of the subendometrial vascularization flow index by three-dimensional ultransound may beuseful for predicting the pregnancy rate for patient undergoing in vitro fertklization embryo transfer.Fertil Steril,2003,79:507~511
    62 Alcazar JL, Three-dimensional ultrasound assessment of endometrial receptivity: a review. Reprod Biol Endocrinol,2006,4:56
    63 Merce LT, Barco MJ, Bau S,et al. Are endometrial parameters by three-dimensional ultrasound and power Doppler angiography related to in vitro fertilization/embryo transfer outcome?Fertil Steril,2008,89:111~117
    64 Ng EH, Chan CC, Tang OS, et al. The role of endometrial and subendometrial blood flows measured by three-dimensional power Doppler ultrasound in the prediction of pregnancy during in vitro fertilization treatment. Hum Reprod,2007,135:8~16
    65 Raga F, Bonilla-Musoles F, Casan EM, et al. Assessment of endometrial volume by three-dimensional ultrasound prior to embryo transfer: clues to endometrial receptivity. Hum Reprod,1999,14:2851~2854
    66 Martins Wde P, Ferriani RA, Nastri CO et al. Measurement of endometrial volume increase during the first week after embryo transfer by three dimensional ultrasound to detect pregnancy:a preliminary study.Fertil Steril,2008,90:883~885
    67 Sozen I, Arici A. Interactions of cytokines, growth factors, and the extracellularmatrix in the cellular biology of uterine leiomyomata. Fertil Steril,2002,78:1~12
    68 Cook JD, Walker CL. Treatment strategies for uterine leiomyoma: the role of hormonal modulation. Semin Reprod Med,2004,22:105~111
    69 Donnez J, Jadoul P. What are the implications of myomas on fertility?A need for a debate? Hum Reprod,2002,17:1424~1430
    70 Ramzy AM, Sattar M, Amin Y, et al. Uterine myomata and outcome of assisted reproductio. Hum Reprod,1998,13:198~202
    71 Pritts EA. Fibroids and infertility: a systematic review of the evidence. Obstet Gynecol Surv,2001,56:483~491
    72 Surrey ES, Minjarez DA, Stevens JM, et al. Effect of myomectomy on the outcome of assisted reproductive technologies. Fertil Steril,2005,83:1473~1479
    73 Hart R, Khalaf Y, Yeong CT, et al. A prospective controlled study of the effect of intramural uterine fibroids on the outcome of assisted conception. Hum Reprod,2001,16:2411~2417
    74 Surrey ES, Lietz AK, Schoolcraft WB. Impact of intramural leiomyomata in patients with a normal endometrial cavity on in vitro fertilization–embryo transfer cycle outcome. Fertil Steril,2001,75: 405~410
    75 Feinberg EC, Larsen FW, Catherino WH, et al.Comparison of assisted reproductive technology utilization and outcomes between caucasian and African American patients in an equal-access-to-care setting. Fertil Steril, 2006,85:888~894
    76 Levens ED,Stegmann BJ,Feinberg EC,et al. Ultrasonographic characteristics of the endometrium among patients with fibroids undergoing ART.Ferti and Steril,2008,89:1005~1007
    77 Higon MA, Horcajadas JA, Martinez-Conejero JA, et al. Endometrial receptivity and implantation are not affected by the presence of uterine intramural leiomyomas:a clinical and functional genomics analysis.J Clin Endocrinol Metab,2008,93:3490~3498
    78 Taylor HS, Rackow BW. Submucosal uterine leiomyomas have a globe effect on molecular determinants of endometrial receptivity. Fertil Steril, 2008,12(Epub ahead of print)
    79 Buyalos R, Agarwal SK. Endometriosis associated infertility. Curr Opin Obstet Gynecol,2000,12:377~381
    80 Arici A, Oral E, Bukulmez O, et al. The effect of endometriosis on im plantation: results from the Yale University in vitro fertilization and embryo transfer program. Fertil Steril,1996,65:603~607
    81 Lessey BA, Castelbaum AJ, Buck CA,et al. Further characterization of endometrial integrins during the menstrual cycle and in pregnancy. Fertil Steril,1994,62:497~506
    82 Lessey BA, Castelbaum AJ, Sawin SW, et al. Aberrant integrin expressionin the endometrium of women with endometriosis. J Clin Endocrinol Metab,1994,79:643~649
    83 Creus M, Balasch J, Ordi J, et al. Integrin expression in normal and out-of-phase endometria. Hum Reprod,1998,13:3460~3468
    84 Ordi J, Creus M, Casamitjana R, et al. Endometrial pinopode and alphavbeta3 integrin expression is not impaired in infertile patientswith endometriosis. J Assist Reprod Genet,2003,20:465~473
    85 Garcia-Velasco JA, Nikas G, Remohi J, et al. Endometrial receptivity in terms of pinopode expression is not impaired in women with endometriosis in artificially prepared cycles. Fertil Steril,2001,75:1231~1233
    86 Diaz I, Navarro J, Blasco L et al. Impact of stage III and IV endometriosis on recipients of sibling oocytes: Matched case - control study. Fertil Steril, 2000,74:31~34
    87 Dimitriadis E, Stoikos C, Stafford-Bell M, et al. Interleukin-11, IL- 11 receptoralpha and leukemia inhibitory factor are dysregulated in endometrium of infertile women with endometriosis during the implantation window. J Reprod Immunol,2006,69:53~64
    88 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,2007,148:3814~3826
    89 Vandromme J, Chasse E, Lejeune B, et al. Hydrosalpinges in in-vitro fertilization: an unfavourable prognostic feature. Hum Reprod, 1995,10:576~579
    90 Fleming C, Hull MG. Impaired implantation after in vitro fertilisation treatment associated with hydrosalpinx. Br J Obstet Gynaecol,1996, 103:268 ~272
    91 Nackley AC, Muasher SJ. The significance of hydrosalpinx in in vitro fertilization. Fertil Steril,1998,69:373~384
    92 Mansour RT, Aboulghar MA, Serour GI, et al. Fluid accumulation of the uterine cavity before embryo transfer: a possible hindrance for implantation. J In Vitro Fert Embryo Transf,1991,8:157~159
    93 Sharara FI. The role of hydrosalpinx in IVF: simply mechanical? Hum Reprod,1999,14:577~578
    94 Daftary GS, Taylor HS. Hydrosalpinx fluid diminishes endometrial cell HOXA10 expression. Fertil Steril,2002,78:577~580
    95 Daftary GS, Kayisli U, Seli E, et al. Salpingectomy increases peri-implantation endometrial HOXA10 expression in women with hydrosalpinx. Fertil Steril,2007,87:367~372
    96 Seli E, Kayisli UA, Cakmak H, et al. Removal of hydrosalpinges increases endometrial leukaemia inhibitory factor(LIF) expression at the time of the implantation window.Hum Reprod,2005,20:3012~3017
    97李艳萍,赵红翠输卵管积水对窗口期子宫内膜基因谱的影响,生命科学研究,2007,11(4):367~372
    98 Lessey BA, Castelbaum AJ, Wolf L, et al. Use of integrins todate the endometrium. Fertil Steril,2000,73:779~787
    99 Bildirici I, Bukulmez O, Ensari A, et al. Aprospective evaluation of the effect of salpingectomy onendometrial receptivity in cases of women with communicating hydrosalpinges. Hum Reprod,2001,16:2422~2426
    100 Savaris RF, Pedrini JL, Flores R,et al. Expression of alpha 1 and beta 3 integrins subunits in the endometrium of patients with tubal phimosis or hydrosalpinx.Fertil Steril,2006,85:188~192
    101 Savaris RF, Giudice LC. The Influence of hydrosalpinx on markers of endometrial receptivity.Semi Reprod Med,2002,5:476~482
    102 Ng EH, Chan C, Tang OS, et al. Comparison of endometrial and subendometrial blood flows among patients with and without hudrosalpinx shown on scanning during in vitro fertilization treatment. Fertil Steril, 2006,85:333~338
    103 Lane DE. Polycystic ovary syndrome and its differential diagnosis.Obstet Gynecol,2006,61:125~135
    104 Sharara FI, Lim J, McClamrock HD. Endometrial pattern on the day of oocyte retrieval is more predictive of implantation success than the pattern or thickness on the day of hCG administration. J Assist Reprod Genet,1999,16:523~528
    105 Jarvela IY, Sladkevicius P, Kelly S,et al. Evaluation of endometrial receptivity during in-vitro fertilization using three-dimensional power Doppler ultrasound. Ultrasound Obstet Gynecol,2005,26:765~769
    106 Daftary GS, Taylor HS. Pleiotropic effects of Hoxa10 on the functional development of periimplantation endometrium. Mol Reprod Dev,2004,67: 8~14
    107 Akbas GE, Song J, Taylor HS, et al. A HOXA10 estrogen responseelement( ERE) is differentially regulated by 17 betaestradiol and diethylstilbestrol(DES). J Mol Biol,2004,340:1013~1023
    108 Cermik D, Selam B, Taylor HS, et al. Regulation of HOXA 10 expression by testosterone in vitro and in the endometrium of patients withpolycystic ovary syndrome . Clin Endocrinol Metab,2003,88:238~243
    109 Apparao KB,Lovely LP,Gui Y,et al. Impaered uterine perfusion associated with metabolic disorders in women with polycystic ovary synduome. Acta Obtet Gynecol Scand,2005,84:189~195
    110 Quezada S, Avellaira C, Johnson MC, et al. Evaluation of steroid receptors ,voregulators ,and molecules associated with uterine receptivity in secretory endometria from untreated women with polycystic ovary syndrome. Fertil Steril,2006,85:1017~1026
    111 Chekir C, Nakatsuka M, Kamada Y, et al. Impaered uterine perfusion associated with metabolic disorders in women with polycystic ovary synduome. Acta Obtet Gynecol Scand,2005,84:189~195

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700