Endometriosis: Pathogenesis and Treatment 2014 Ed.

16. Potential New Drugs for Endometriosis: Experimental Evidence

Kaei Nasu Yukie Kawano1Masakazu Nishida1Akitoshi Tsuno1Akitoshi Yuge1Wakana Abe1Kentaro Kai1Mamiko Okamoto1 and Hisasshi Narahara1

(1)

Department of Obstetrics and Gynecology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama-machi, Yufu-shi Oita, 879-5593, Japan

Kaei Nasu

Email: nasu@oita-u.ac.jp

Abstract

Endometriosis, a disease affecting 3–10 % of women of reproductive age, is characterized by the ectopic growth of endometrial tissue. Recent basic studies have revealed that the dysregulation of apoptosis, fibrosis, and epigenetic factors plays important roles in the pathogenesis of this enigmatic disease.

Contraceptive steroids, progestogens, agonists of gonadotropin-releasing hormone, androgens, and nonsteroidal anti-inflammatory agents have been used to treat endometriosis. Endometriosis treatments designed to lower circulating estradiol concentrations can be used only for a limited time due to unacceptable side effects. The development of medical treatments based on novel strategies to prevent or treat endometriosis has thus become a research priority.

Regarding the development of novel medical treatments for endometriosis, many researchers have been evaluating new drugs including molecular-targeting agents and herbal medicine as well as the newly developed hormonal agents. This chapter is a review of the findings from recent basic research on the pathogenesis of endometriosis and the evaluations of novel medical treatments for this disease, especially focusing on the inhibitors of nuclear factor-κB, the mevalonate-Rho/ROCK pathway, and histone deacetylase. These agents are now considered promising agents for the treatment and prevention of endometriosis.

Keywords

EndometriosisHistone deacetylase inhibitorMevalonate-Rho/ROCK pathwayNuclear factor-κB

16.1 Introduction

Endometriosis—the benign, estrogen-dependent, tumorlike disease characterized by chronic pelvic pain, dysmenorrhea, dyspareunia, and/or subfertility—is due to the uncontrolled ectopic growth of proliferative endometrial tissue. Women of reproductive age are most commonly affected by endometriosis, which usually occurs in the peritoneum, ovaries, and rectovaginal septum [1]. The symptoms of endometriosis may markedly reduce a woman’s quality of life.

Several surgical and medical strategies have been used to treat endometriosis. Contraceptive steroids, progestogens, and agonists of gonadotropin-releasing hormone, androgens, and nonsteroidal anti-inflammatory agents have been attempted [2]. Endometriosis treatments designed to lower circulating estradiol concentrations can be used only for a limited time due to unacceptable side effects. The current medical treatments inhibit the growth of endometriotic implants by suppressing ovarian steroids and inducing a hypoestrogenic state; they have been demonstrated to be effective in relieving endometriosis-associated pain [2]. However, high recurrence rates, up to 45 %, after the completion of medical treatments remain a significant problem [3]. An international consensus group proposed the development of nonhormonal medical treatments to prevent or treat endometriosis as a research priority [4]. In addition, many research groups have been evaluating new drugs for endometriosis such as molecular-targeting agents and herbal medicine, as well as the newly developed hormonal agents.

In this chapter, we review the recent basic studies on the development of novel medical treatments for endometriosis based on distinct strategies. We focus on the recent publications about the nuclear factor (NF)-κB inhibitors, mevalonate-Ras homology (Rho)/Rho-associated coiled-coil-forming protein kinase (ROCK) inhibitors, and histone deacetylase (HDAC) inhibitors (HDACIs) as potential candidates for the next era in endometriosis treatment and prevention.

16.2 Potential New Drugs

16.2.1 NF-κB Inhibitors

Apoptosis plays a critical role in maintaining tissue homeostasis, and its function is to eliminate excess cells and dysfunctional cells. Apoptosis can be initiated by extracellular or intracellular “death signals,” and it results from a series of related morphologic and biochemical processes. Morphologically, apoptotic cells present with condensed chromatin, multiple membrane-bound organelles (apoptotic bodies), and shrunken appearance. Biochemically, apoptosis is characterized by monomeric or multimeric 180-base pair nucleosomal fragments resulting from the cleavage of double-stranded nuclear DNA [5]. Apoptosis is controlled by the expression of a number of regulatory genes, including c-myc, p53, Fas, NF-κB, and members of the B-cell lymphoma/leukemia (Bcl)-2 family [611].

Histologically, endometriotic tissues and normal proliferative endometrium are similar. However, endometriosis is increasingly being recognized as a condition in which endometriotic cells at the ectopic sites exhibit abnormal proliferative and apoptotic regulation in response to appropriate stimuli [1222]. It has been demonstrated that the degree of apoptosis in endometriotic lesions is less than that in the endometrium of the same patients and that of healthy women [13152325]. The resistance of endometriotic cells to apoptosis is suspected to be either intrinsic or brought about by environmental factors. Aberrantly expressed proliferation-related and apoptosis-related molecules in endometriosis include the decreased expressions of homeobox (HOX) A10 [26] and caspase-1 [27] and the enhanced expressions of c-myc [28], cellular inhibitor of apoptosis protein (cIAP) 1 [29], X chromosome-linked IAP [29], B-cell lymphoma/leukemia (Bcl)-2 [19], Bcl-XL [19], and NF-κB [3031] in endometriotic cells.

The pleiotropic transcription factor NF-κB has been identified as a critical component of several signal transduction pathways [29]. Figure 16.1 shows the proposed functions of NF-κB in the pathogenesis of endometriosis. One important function of NF-κB is its ability to protect cells from apoptosis by activating antiapoptotic genes [910]. Wieser et al. [32] demonstrated the constitutive activation of NF-κB in endometriotic cells. It is suggested that NF-κB has a significant role in the proliferation of endometriotic lesions [3031]. The activation of NF-κB by lipopolysaccharide (LPS) induces the proliferation of endometriotic stromal cells [33].

A313895_1_En_16_Fig1_HTML.gif

Fig. 16.1

Involvement of the NF-κB-mediated pathway in the pathogenesis of endometriosis. ECM extracellular matrix, IκB inhibitor κB, IKK IκB kinase, NIK NF-κB-inducing kinase, MAPKKMAPK kinase, TNF tumor necrosis factor

A number of substances have been presented in the endometriosis literature as NF-κB inhibitors. The reported selective inhibitors of NF-κB include an IκB protease inhibitor, N-tosyl-L-phenylalanine chloromethyl ketone (TPCK) [33], thalidomide [34], BAY 11–7085 [35], pyrrolidine dithiocarbamate (PDTC) [3638], costunolide [39], and parthenolide [40]. The urinary preparation human chorionic gonadotropin A (hCG-A) [41], gonadotropin-releasing hormone (GnRH) agonists [30], progesterone [42], and danazol [42] have been demonstrated to inhibit NF-κB activity in endometriosis. NF-κB inhibitors have been shown to significantly block the proliferation of endometriotic stromal cells [333540] and they induce apoptosis and the G0/G1 phase cell-cycle arrest of endometriotic stromal cells [353839]. Additionally, after endometriotic stromal cells were treated with NF-κB inhibitors, the downregulation of the expression of antiapoptotic factors (Bcl-2 and Bcl-XL), inflammatory cytokines (interleukin-6 [IL-6], IL-8, monocyte chemoattractant protein-1 [MCP-1], and granulocyte-macrophage colony-stimulating factor [GM-CSF]), inflammatory mediators (COX-2 and PGE2), extracellular matrix remodeling mediators (MMP-2, MMP-3, MMP-7, MMP-9), CD44, and vascular endothelial growth factor [VEGF] with simultaneous activation of caspase-3, caspase-8, and caspase-9 was observed [303441].

The suppression of NF-κB activity by proteasome inhibitors also suppresses the proliferation of endometriotic cells in vitro [31]. The NF-κB inhibitors BAY 11–7085 and SN-50 significantly reduced the development of endometriotic lesions in a nude mice model [43]. Takai et al. [40] also demonstrated that parthenolide reduced the growth of endometriotic lesions in a murine model. The antioxidant PDTC reduced the growth and vascularity of experimental peritoneal endometriotic lesions in a rat model [44].

16.2.2 Mevalonate-Rho/ROCK Inhibitors

During the development and progression of endometriotic lesions, excess fibrosis may lead to scarring, chronic pain, and the alterations of tissue function that are characteristic of this disease [4548]. It has been suggested that type I collagen is a major contributor to endometriosis-associated fibrosis [4649], whereas α-smooth muscle actin (SMA)-positive fibroblastic cells were frequently detected in the fibrotic areas associated with endometriosis of the peritoneum, ovary, rectovaginal septum, and uterosacral ligaments [454850]. An immunohistochemical analysis led Anaf et al. [50] to suggest that endometriotic stromal cells can differentiate to α-SMA-positive myofibroblasts.

To establish a model of fibrosis formation in endometriosis, we used a 3D collagen culture system with human endometriotic stromal cells [5152]. We cultured the cells in floating collagen lattices to reorganize the collagen fibers and make them compact, resulting in contraction of the collagen gels. This culture system provided a model for mechanically relaxed tissue with low tensile strength, comparable to the early stages of an endometriotic lesion. We found that endometriotic stromal cells cultured in floating 3D gels have an enhanced contractile profile compared to normal endometrial stromal cells [51]. This suggested that endometriotic stromal cells may acquire fibrogenetic ability or fail to avoid fibrogenesis during the pathogenesis of endometriosis.

Members of the Rho family of small guanosine triphosphatase (GTPase) are known to regulate cell shape, motility, cell-substratum adhesion, and cell contraction through the reorganization of actin cytoskeletons [5365]. The active form of Rho is GTP-bound [5462], and many polypeptides have been reported as targets of activated Rho, including ROCK-I/p160ROCK and Rho-kinase/ROCK-II [66]. ROCKs phosphorylate the myosin light chain (MLC) [67] and the myosin-binding subunit of myosin phosphatase [68], and they inhibit myosin II regulatory light chain phosphatase activity [68] in cultured fibroblasts. Rho and ROCKs have been implicated in myosin II-dependent force generation [69]. Thus, the activation of ROCKs by Rho can promote the assembly of focal adhesions, actin stress fiber formation, and contraction of non-muscle cells [6268], in which RhoA regulates α-SMA expression [70].

Based on these observations in fibroblasts, we have investigated the signaling pathway underlying endometriotic stromal cell-mediated contractility by using 3D cultures. Our data indicated that human endometriotic stromal cells undergo myofibroblastic differentiation and show increased expression of RhoA, ROCK-I, and ROCK-II proteins, resulting in enhanced contractility [51]. Thus, we evaluated several inhibitors of mevalonate-Rho/ROCK pathways as candidate drugs for the treatment and prevention of endometriosis-associated fibrosis [51527173].

16.2.3 Statins

Statins are potent inhibitors of cholesterol biosynthesis that are widely used to reduce serum cholesterol levels in hyperlipidemic patients [7475]. Statins are divided into three categories: natural statins (i.e., lovastatin and pravastatin), semisynthetic statins (i.e., simvastatin), and synthetic statins (i.e., atorvastatin, fluvastatin, and cerivastatin) [7576]. These three subtypes of statin exhibit markedly different hydrophobicities, with simvastatin as the most hydrophobic and pravastatin as the most hydrophilic.

Statins competitively inhibit 3-hydroxy-3-methylglutarylcoenzyme A (HMG-CoA) reductase to block the conversion of HMG-CoA to L-mevalonate, a rate-limiting step in cholesterol synthesis [77]. By inhibiting the initial step of the cholesterol synthesis pathway, statins reduce the synthesis of several important lipid intermediate compounds including isoprenoids such as farnesyl pyrophosphate (FPP), a precursor of cholesterol, and geranylgeranyl pyrophosphate (GGPP), which is synthesized from FPP [78]. Both FPP and GGPP are important isoprenoid intermediates and serve as lipid attachments for a variety of intercellular proteins to the plasma membrane, including Rho proteins, resulting in their activation [5979]. By inhibiting the activation of Rho proteins, statins also regulate both the Rho/ROCK pathways and the mevalonate pathway [8081].

Simvastatin has been demonstrated to inhibit the proliferation of endometriotic stromal cells as well as the collagen gel contraction mediated by these cells [71]. Attenuation of the endometriotic stromal cell attachment to collagen fibers is involved in this mechanism. Lovastatin also inhibited cell proliferation and angiogenesis in endometriosis [82], whereas simvastatin and mevastatin inhibited the MCP-1 production by endometriotic cells [83]. Atorvastatin also exhibited antiproliferative and anti-inflammatory effects in endometriotic cells [84]. Oktem et al. [85] demonstrated that atorvastatin induced the regression of endometriotic implants in a rat model. Similarly, Bruner-Tran et al. [86] demonstrated that simvastatin induced the regression of endometriotic implants in a nude mouse model.

16.2.4 ROCK Inhibitors

Several selective ROCK inhibitors including Y-27632 and fasudil hydrochloride are now available for basic and clinical studies [58638788]. We demonstrated that Y-27632, a pyridine derivative that acts as a specific inhibitor of ROCK-I and ROCK-II [5863], inhibits endometriotic stromal cell-mediated contractility [51]. Interestingly, Y-27632 was found to exert a greater effect on the contractility of endometriotic stromal cells than on the contractility of normal eutopic endometrial stromal cells, whereas fasudil hydrochloride also inhibited endometriotic stromal cell-mediated contractility, myofibroblastic differentiation, and cell proliferation [72].

16.2.5 Heparin

Heparin is an analog of heparan sulfate, a unique class of macromolecules that is widely expressed on the cell surface and in the extracellular matrix [89]. It is a commonly used anticoagulant drug [90]. Heparin was shown to inhibit the gel contraction mediated by dermal fibroblasts [9192]. We demonstrated that heparin attenuates the contractility of endometriotic stromal cells by suppressing the cells’ attachment to collagen fibers, the inhibition of myofibroblastic differentiation, and the suppression of the Rho/ROCK-mediated pathway [73]. However, the precise target molecule of heparin’s action on endometriotic stromal cells has not yet been elucidated.

16.2.6 HDACIs

Epigenetics refers to the stable inheritance of phenotypes of cells and organisms without changes in DNA sequence or DNA content [93]. The epigenetic phenotypes are conferred via nuclear processes such as DNA methylation and chromatin modifications (e.g., acetylation, biotinylation, isomerization, methylation, phosphorylation, ribosylation, sumoylation, and ubiquitination of histones) and underlie the regulation of all genome functions, including gene expression, DNA replication, and genome stability [9496]. Epigenetic phenotypes are also conferred via microRNA and double-stranded noncoding RNA, which are interconnected and may work together to establish and/or maintain specific gene activity states in normal cells [939697]. Epigenetic processes are known to be involved in development, health, disease, and aging and are responsible for phenomena such as X-chromosome inactivation and genomic imprinting [9899]. Of these epigenetic regulatory mechanisms, histone acetylation is the most studied.

The acetylation levels of histone are controlled by a balance between histone acetyltransferases and histone deacetylases (HDACs). Histone acetyltransferases transfer acetyl groups from acetyl-CoA to lysine residues on the aminoterminal region of histones and activate genetic transcription. Conversely, HDACs restore the positive charge on lysine residues (by removing the acetyl groups) and prevent transcription. HDACs are large multiprotein complexes that target promoter sites through their interaction with sequence-specific transcription. They play an important role in the regulation of gene transcription through the remodeling of chromatin structure and dynamic changes in the nucleosomal packaging of DNA [100].

The hyperacetylation of histones H3 and H4 is often associated with activated transcription, and the hypoacetylation of histones H3 and H4 correlates with transcriptional silencing or repression [101], whereas Choi et al. [102] suggested that the substrates of HDACs are not restricted to histones but include transcriptional regulators, such as p53, E2F-1, Mad-1, BCL-6, and ETO. In this regard, global gene expression analyses have shown that HDACIs affect the expression levels of 2–20 % of genes in the genome, of which about half are upregulated and half downregulated [103].

Accumulating evidence indicates that several epigenetic aberrations are involved in the pathogenesis of endometriosis [104110]. We observed that the levels of acetylated histones H3 and H4 were significantly lower in unstimulated endometriotic stromal cells compared to normal endometrial stromal cells, suggesting that aberrant histone modifications are present in endometriosis [108]. This initial finding encouraged us to evaluate the efficacy of HDACIs for the treatment of endometriosis. Our subsequent experiments demonstrated that HDACIs significantly inhibited the proliferation of endometriotic stromal cells, and they also induced significant levels of cell-cycle arrest at the G0/G1 or G2/M phases and significant apoptosis of these cells [108].

Interestingly, HDACIs showed marginal to weak effects on normal endometrial stromal cells compared to endometriotic stromal cells. Moreover, HDACI treatment significantly inhibited HDAC activity and resulted in the accumulation of acetylated histones H3 and H4 in total cellular chromatin and in the promoter regions of the p16INK4a, p21Waf1/Cip1, p27Kip1, and cell-cycle checkpoint kinase 2 (chk2) genes in these cells. A Western blot analysis revealed the increased protein levels of p16INK4a, p21Waf1/Cip1, p27Kip1, and chk2, the suppression of Bcl-2 and Bcl-XL protein levels, and the activation of caspase-3 and caspase-9 in endometriotic stromal cells after treatment with HDACIs [108]. Recently, we found that the expression of CCAAT/enhancer-binding protein (C/EBP)-α, a tumor suppressor gene, and death receptor 6, a TNF receptor superfamily gene, was epigenetically suppressed by histone deacetylation (our unpublished data).

Histone deacetylation appears to be a potent regulator of gene expression in endometriosis, which raises the prospect of using HDACIs as therapeutic tools in endometriosis [108]. Several classes of HDACIs have been identified, including (a) organic hydroxamic acids [e.g., trichostatin A and suberoylanilide bishydroxamine (SAHA)], (b) short-chain fatty acids [e.g., butyrates and valproic acid (VPA)], (c) benzamides (e.g., MS-275), (d) cyclic tetrapeptides (e.g., trapoxin), and (e) sulfonamide anilides [108]. The molecular events that mediate the biological effects of HDACIs are incompletely understood. Inhibition of HDAC by HDACIs increases histone acetylation and maintains chromatin structure in a more open conformation, resulting in the reactivation of transcriptionally silenced pathways or the suppression of aberrantly expressed genes through the recruitment of repressors [111112]. HDACIs can reactivate genes silenced by promoter hypermethylation as well as the demethylation agents [113]. HDACIs also cause mitotic defects through non-transcriptional mechanisms [114].

We demonstrated that HDACIs, including VPA, SAHA, and apicidin, can inhibit the proliferation, induce cell differentiation and cell-cycle arrest, and stimulate the apoptosis of endometriotic cells [108]. Guo and his colleagues have demonstrated that HDACIs, such as VPA and trichostatin A, can suppress the proliferation, induce cell-cycle arrest, inhibit IL-1β-induced cyclooxygenase-2 expression and NF-κB activation, upregulate peroxisome proliferator-activated receptor γ, p21Waf1/Cip1 and PR-B expression, attenuate invasiveness, and reactivate the silenced E-cadherin gene expression of endometriotic cells [115120]. Romidepsin, an HDACI, was shown to specifically reduce HDAC enzymatic activity, resulting in inhibited cell proliferation, cell-cycle arrest, increased apoptosis, and reduced expression of VEGF mRNA and protein in endometriotic cells [121122]. Treatment with trichostatin A and VPA has been shown to significantly reduce the growth of endometriotic lesions in a murine model [123124]. Histone deacetylation appears to be a potent regulator of gene expression in endometriosis, which raises the prospect of using HDACIs as therapeutic tools in endometriosis [108117].

VPA was used in a pilot study of three patients with endometriosis and adenomyosis who had moderate to severe dysmenorrhea [125]. A VPA dose of 1,000 mg/day was used for 3 months. Complete relief of pain in all cases and an average reduction of one-third in uterine size were reported. The disappearance or reduction of palpable tender nodules in the cul-de-sac was also reported.

16.3 Conclusions

Basic research on endometriosis offers new opportunities to understand the pathogenesis of this enigmatic disease and to provide novel medical treatments other than hormonal therapy. As shown in this chapter, NF-κB inhibitors and mevalonate-Rho/ROCK inhibitors as well as HDACIs seem to be promising therapeutic strategies for the treatment of endometriosis.

Acknowledgments

This work was supported in part by a Grant-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (no. 13237327 to K. Nasu, no. 25861500 to Y. Kawano, and no. 23592407 to H. Narahara).

References

1.

Giudice LC, Kao LC. Endometriosis. Lancet. 2004;364:1789–99.PubMed

2.

Practice Committee of the American Society for Reproductive Medicine. Endometriosis and infertility. Fertil Steril. 2004;81:1441–6.

3.

Bergqvist A. A comparative study of the acceptability and effect of goserelin and nafarelin on endometriosis. Gynecol Endocrinol. 2000;14:425–32.PubMed

4.

Rogers PA, D’Hooghe TM, Fazleabas A, Gargett CE, Giudice LC, Montgomery GW, Rombauts L, Salamonsen LA, Zondervan KT. Priorities for endometriosis research: recommendations from an international consensus workshop. Reprod Sci. 2009;16:335–46.PubMedCentralPubMed

5.

Oberhammer F, Wilson JW, Dive C, et al. Apoptotic death in epithelial cells: cleavage of DNA to 300 and/or 50 kb fragments prior to or in the absence of internucleosomal fragmentation. EMBO J. 1993;12:3679–84.PubMedCentralPubMed

6.

White E. Death-defying acts: a meeting review on apoptosis. Genes Dev. 1993;7:2277–84.PubMed

7.

Osborne BA, Schwartz LM. Essential genes that regulate apoptosis. Trends Cell Biol. 1994;4:394–8.PubMed

8.

Nagata S, Golstein P. The Fas death factor. Science. 1995;267:1449–56.PubMed

9.

Beg AA, Baltimore D. An essential role for NF-κB in preventing TNF-α-induced cell death. Science. 1996;274:782–4.PubMed

10.

Van Antwerp DJ, Martin SJ, Kafri T, Green DR, Verma IM. Suppression of TNF-α-induced apoptosis by NF-κB. Science. 1996;274:787–9.PubMed

11.

Sattler M, Liang H, Nettesheim D, et al. Structure of Bcl-xL-BaK peptide complex: recognition between regulators of apoptosis. Science. 1997;225:983–6.

12.

Harada M, Suganuma N, Furuhashi M, Nagasaka T, Nakashima N, Kikkawa F, Tomoda Y, Furui K. Detection of apoptosis in human endometriotic tissues. Mol Hum Reprod. 1996;2:307–15.PubMed

13.

Dmowski WP, Gebel H, Braun DP. Decreased apoptosis and sensitivity to macrophage mediated cytolysis of endometrial cells in endometriosis. Hum Reprod Update. 1998;4:696–701.PubMed

14.

Gebel HM, Braun DP, Tambur A, Frame D, Rana N, Dmowski WP. Spontaneous apoptosis of endometrial tissue is impaired with endometriosis. Fertil Steril. 1998;69:1042–7.PubMed

15.

Imai A, Takagi A, Tamaya T. Gonadotropin-releasing hormone analog repairs reduced endometrial cell apoptosis in endometriosis in vitro. Am J Obstet Gynecol. 2000;182:1142–6.PubMed

16.

Selam B, Arici A. Implantation defect in endometriosis: endometrium or peritoneal fluid. J Reprod Fertil Suppl. 2000;55:121–8.PubMed

17.

Peiro G, Diebold J, Buretton GB, Kimmig R, Lohrs U. Cellular apoptosis susceptibility gene expression in endometrial carcinoma: correlation with Bcl-2, Bax, and Caspase-3 expression and outcome. Int J Gynecol Pathol. 2001;20:359–67.PubMed

18.

Harada T, Kaponis A, Iwabe T, Taniguchi F, Makrydimas G, Sofikitis N, et al. Apoptosis in human endometrium and endometriosis. Hum Reprod Update. 2004;10:29–38.PubMed

19.

Nishida M, Nasu K, Ueda T, Fukuda J, Takai N, Miyakawa I. Endometriotic cells are resistant to interferon-γ-induced cell growth inhibition and apoptosis: a possible mechanism involved in the pathogenesis of endometriosis. Mol Hum Reprod. 2005;11:29–34.PubMed

20.

Dufournet C, Uzan C, Fauvet R, Cortez A, Siffroi J-P, Daria E. Expression of apoptosis-related proteins in peritoneal, ovarian and colorectal endometriosis. J Reprod Immunol. 2006;70:151–62.PubMed

21.

Izawa M, Harada T, Deura I, Taniguchi F, Iwabe T, Terakawa N. Drug-induced apoptosis was markedly attenuated in endometriotic stromal cells. Hum Reprod. 2006;21:600–4.PubMed

22.

Klemmt PAB, Carver JG, Kennedy SH, Koninckx PR, Mardon HJ. Stromal cells from endometriotic lesions and endometrium from women with endometriosis have reduced decidualization capacity. Fertil Steril. 2006;85:564–72.PubMedCentralPubMed

23.

Suganuma N, Harada M, Furuhashi M, Nawa A, Kikkawa F. Apoptosis in human and endometriotic tissues. Horm Res. 1997;48:42–7.PubMed

24.

Jones RK, Searle RF, Bulmer JN. Apoptosis and bcl-2 expression in normal human endometrium, endometriosis and adenomyosis. Hum Reprod. 1998;13:3496–502.PubMed

25.

Beliard A, Noel A, Foidart J-M. Reduction of apoptosis and proliferation in endometriosis. Fertil Steril. 2004;82:80–5.PubMed

26.

Taylor HS, Bagot C, Kardana A, Olive D, Arici A. HOX gene expression is altered in the endometrium of women with endometriosis. Hum Reprod. 1999;14:1328–31.PubMed

27.

Braun DP, Ding J, Shaheen F, Willey JC, Rana N, Dmowski WP. Quantitative expression of apoptosis-regulating genes in endometrium from women with and without endometriosis. Fertil Steril. 2007;87:263–8.PubMed

28.

Pellegrini C, Gori I, Achtari C, Hornung D, Chardonnens E, Wunder D, Fiche M, Canny GO. The expression of estrogen receptors as well as GREB1, c-MYC, and Cyclin D1, estrogen-regulated genes implicated in proliferation, is increased in peritoneal endometriosis. Fertil Steril. 2012;98:1200–8.PubMed

29.

Watanabe A, Taniguchi F, Izawa M, Suou K, Uegaki T, Takai E, et al. The role of survivin in the resistance of endometriotic stromal cells to drug-induced apoptosis. Hum Reprod. 2009;24:3172–9.PubMed

30.

Sakamoto Y, Harada T, Horie S, Iba Y, Taniguchi F, Yoshida S, et al. Tumor necrosis factor-a-induced interleukin-8 (IL-8) expression in endometriotic stromal cells, probably through nuclear factor-κB activation: gonadotropin releasing hormone agonist treatment reduced IL-8 expression. J Clin Endocrinol Metab. 2003;88:730–5.PubMed

31.

Guo S-W. Nuclear factor-κB (NF-κB): an unsuspected major culprit in the pathogenesis of endometriosis that is still at large? Gynecol Obstet Invest. 2007;63:71–97.PubMed

32.

Wieser F, Vigne J-L, Ryan I, Hornung D, Djalali S, Taylor RN. Sulindac suppresses nuclear factor-κB activation and RANTES gene and protein expression in endometrial stromal cells from women with endometriosis. J Clin Endocrinol Metab. 2005;90:6441–7.PubMed

33.

Iba Y, Harada T, Horie S, Deura I, Iwabe T, Terakawa N. Lipopolysaccharide-promoted proliferation of endometriotic stromal cells via induction of tumor necrosis factor a and interleukin-8 expression. Fertil Steril. 2004;82 Suppl 3:1036–42.PubMed

34.

Yagyu T, Kobayashi H, Matsuzaki H, Wakahara K, Kondo T, Kurita N, et al. Thalidomide inhibits tumor necrosis factor-a-induced interleukin-8 expression in endometriotic stromal cells, possibly through suppression of nuclear factor-κB activation. J Clin Endocrinol Metab. 2005;90:3017–21.PubMed

35.

Nasu K, Nishida M, Ueda T, Yuge A, Takai N, Narahara H. Application of the selective nuclear factor-κB inhibitor, BAY 11–7085, for the treatment of endometriosis: an in vitro study. Am J Physiol Endocrinol Metab. 2007;293:E16–23.PubMed

36.

Zhang JJ, Xu ZM, Dai HY, Ji XQ, Duan YY, Zhang CM, et al. Application of the nuclear factor-κB inhibitor pyrrolidine dithiocarbamate for the treatment of endometriosis: an in vitro study. Fertil Steril. 2010;94:2942–4.PubMed

37.

Zhang HI, Li M, Wang F, Liu S, Li J, Wen Z, et al. Endometriotic epithelial cells induce MMPs expression in endometrial stromal cells via an NFқB-dependent pathway. Gynecol Endocrinol. 2010;26:456–67.PubMed

38.

Zhang JJ, Xu ZM, Zhang CM, Dai HY, Ji XQ, Wang XF, et al. Pyrrolidine dithiocarbamate inhibits nuclear factor-κB pathway activation and regulates adhesion, migration, invasion and apoptosis of endometriotic stromal cells. Mol Hum Reprod. 2011;17:175–81.PubMed

39.

Kim JH, Yang YI, Lee KT, Park HJ, Choi JH. Costunolide induces apoptosis in human endometriotic cells through inhibition of the prosurvival Akt and nuclear factor kappa B signaling pathway. Biol Pharm Bull. 2011;34:580–5.PubMed

40.

Takai E, Taniguchi F, Uegaki T, Iwabe T, Terakawa N, Harada T. Parthenolide reduces cell proliferation and PGE2 synthesis in human endometriotic stromal cells and inhibits development of endometriosis in murine mode. J Endometriosis. 2012;4:165.

41.

Huber AV, Saleh L, Prast J, Haslinger P, Knöfler M. Human chorionic gonadotropin attenuates NF-κB activation and cytokine expression of endometriotic stromal cells. Mol Hum Reprod. 2007;13:595–604.PubMed

42.

Horie S, Harada T, Mitsunari M, Taniguchi F, Iwabe T, Terakawa N. Progesterone and progestational compounds attenuate tumor necrosis factor alpha-induced interleukin-8 production via nuclear factor kappaB inactivation in endometriotic stromal cells. Fertil Steril. 2005;83:1530–5.PubMed

43.

Gonzalez-Ramos R, Van Langendonckt A, Defrere S, Lousse JC, Mettlen M, Guillet A, Donnez J. Agents blocking the nuclear factor-κB pathway are effective inhibitors of endometriosis in an in vivo experimental model. Gynecol Obstet Invest. 2008;65:174–86.PubMed

44.

Celik O, Hascalik S, Elter K, Tagluk ME, Gurates B, Aydin NE. Combating endometriosis by blocking proteasome and nuclear factor-κB pathways. Hum Reprod. 2008;23:2458–65.PubMed

45.

Nisolle M, Donnez J. Peritoneal endometriosis, ovarian endometriosis, and adenomyotic nodules of the rectovaginal septum are three different entities. Fertil Steril. 1997;68:585–96.PubMed

46.

Matsuzaki S, Canis M, Darcha C, Dechelotte P, Pouly J-L, Bruhat MA. Fibrogenesis in peritoneal endometriosis. Gynecol Obstet Invest. 1998;47:197–9.

47.

Bonte H, Chapron C, Vieira M, Fauconnier A, Barakat H, Fritel X, Vacher-Lavenu M-C, Dubuisson J-B. Histologic appearance of endometriosis infiltrating uterosacral ligaments in women with painful symptoms. J Am Assoc Gynecol Laparosc. 2002;9:519–24.PubMed

48.

Itoga T, Matsumoto T, Takeuchi H, Yamasaki S, Sasahara N, Hoshi T, Kinoshita K. Fibrosis and smooth muscle metaplasia in rectovaginal endometriosis. Pathol Int. 2003;53:371–5.PubMed

49.

Stovall DW, Anners JA, Halme J. Immunohistochemical detection of type I, III, and IV collagen in endometriotic implants. Fertil Steril. 1992;57:984–9.PubMed

50.

Anaf V, Simon P, Fayt I, Noel J-C. Smooth muscles are frequent components of endometriotic lesions. Hum Reprod. 2000;15:767–71.PubMed

51.

Yuge A, Nasu K, Matsumoto H, Nishida M, Narahara H. Collagen gel contractility is enhanced in human endometriotic stromal cells: a possible mechanism underlying the pathogenesis of endometriosis-associated fibrosis. Hum Reprod. 2007;22:938–44.PubMed

52.

Nasu K, Yuge A, Tsuno A, Narahara H. Mevalonate-Ras homology (Rho)/Rho-associated coiled-coil-forming protein kinase (ROCK)-mediated signaling pathway as a therapeutic target for the treatment of endometriosis-associated fibrosis. Curr Signal Transduct Ther. 2010;5:141–8.

53.

Ridley AJ, Hall A. The small GTP-binding protein rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors. Cell. 1992;70:389–99.PubMed

54.

Lee T-L, Lin Y-C, Mochitate K, Grinnell F. Stress-relaxation of fibroblasts in collagen matrices triggers ectocytosis of plasma membrane vesicles containing actin, annexins II and VI, and β1 integrin receptors. J Cell Sci. 1993;105:167–77.PubMed

55.

Amano M, Mukai H, Ono Y, Chihara K, Matsui T, Hamajima Y, Okawa K, Iwamatsu A, Kaibuchi K. Identification of a putative target for Rho as the serine-threonine kinase protein kinase N. Science. 1996;271:648–50.PubMed

56.

Kimura K, Ito M, Amano M, Chihara K, Fukata Y, Nakafuku M, Yamamori B, Feng J, Nakano T, Okawa K, Iwamatsu A, Kaibuchi K. Regulation of myosin phosphatase by Rho and Rho-associated kinase (Rho-kinase). Science. 1996;273:245–8.PubMed

57.

Nakagawa O, Fujisawa K, Ishizaki T, Saito Y, Nakao K, Narumiya S. ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett. 1996;39:189–93.

58.

Uehata M, Ishizaki T, Satoh H, Ono T, Kawahara T, Morishita T, Tamakawa H, Yamagami K, Inui J, Maekawa M, Narumiya S. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature. 1997;389:990–4.PubMed

59.

Van Aelst L, D’Souza-Schorey C. Rho GTPases and signaling networks. Genes Dev. 1997;11:2295–322.PubMed

60.

Hall A. Rho GTPases and the actin cytoskeleton. Science. 1998;279:509–14.PubMed

61.

Park HJ, Galper JB. 3-Hydroxy-3-methylglutaryl CoA reductase inhibitors up-regulate transforming growth factor-beta signaling in cultured heart cells via inhibition of geranylgeranylation of RhoA GTPase. Proc Natl Acad Sci U S A. 1999;96:11525–30.PubMedCentralPubMed

62.

Ravanti L, Heino J, Lopez-Otin C, Kahari VM. Induction of collagenase-3 (MMP-13) expression in human skin fibroblasts by three-dimensional collagen is mediated by p38 mitogen-activated protein kinase. J Biol Chem. 1999;274:2446–55.PubMed

63.

Ishizaki T, Uehata M, Tamechika I, Keel J, Nonomura K, Maekawa M, Narumiya S. Pharmacological properties of Y-27632, a specific inhibitor of rho-associated kinases. Mol Pharmacol. 2000;57:976–83.PubMed

64.

Mack CP, Somlyo AV, Hautmann M, Somlyo AP, Owens GK. Smooth muscle differentiation marker gene expression is regulated by RhoA-mediated actin polymerization. J Biol Chem. 2001;276:341–7.PubMed

65.

Etienne-Manneville S, Hall A. Rho GTPases in cell biology. Nature. 2002;420:629–35.PubMed

66.

Rosenfeldt H, Grinnell F. Fibroblast quiescence and the disruption of ERK signaling in mechanically unloaded collagen matrices. J Biol Chem. 2000;275:3088–92.PubMed

67.

Grundstrom G, Mosher DF, Sakai T, Rubin K. Integrin αvβ3 mediates platelet-derived growth factor-BB-stimulated collagen gel contraction in cells expressing signaling deficient integrin α2β1. Exp Cell Res. 2003;291:463–73.PubMed

68.

Fringer J, Grinnell F. Fibroblast quiescence in floating or released collagen matrices. J Biol Chem. 2001;276:31047–52.PubMed

69.

Carnevali S, Mio T, Adachi Y, Spurzem JR, Striz I, Romberger DJ, Illig M, Rennard SI. Gamma radiation inhibits fibroblast-mediated collagen gel retraction. Tissue Cell. 2003;35:459–69.PubMed

70.

Galois L, Hutasse S, Cortial D, Rousseau CF, Grossin L, Ronziere MC, Herbage D, Freyria AM. Bovine chondrocytes behaviour in three-dimensional type I collagen gel in terms of gel contraction, proliferation and gene expression. Biomaterials. 2006;27:79–90.PubMed

71.

Nasu K, Yuge A, Tsuno A, Narahara H. Simvastatin inhibits the proliferation and the contractility of human endometriotic stromal cells: a promising agent for the treatment of endometriosis. Fertil Steril. 2009;92:2097–9.PubMed

72.

Tsuno A, Nasu K, Kawano Y, Yuge A, Li H, Abe W, Narahara H. Fasudil inhibits the proliferation and contractility and induces cell cycle arrest and apoptosis of human endometriotic stromal cells: a promising agent for the treatment of endometriosis. J Clin Endocrinol Metab. 2011;96:E1944–52.PubMed

73.

Nasu K, Tsuno A, Hirao M, Kobayashi H, Yuge A, Narahara H. Heparin is a promising agent for the treatment of endometriosis-associated fibrosis. Fertil Steril. 2010;94:46–51.PubMed

74.

Molgaard J, Von Schenck H, Olsson AG. Effects of simvastatin on plasma lipid, lipoprotein and apolipoprotein concentrations in hypercholesterolaemia. Eur Heart J. 1988;9:541–51.PubMed

75.

Hamelin BA, Turgeon J. Hydrophilicity/lipophilicity: relevance for the pharmacology and clinical effects of HMG-CoA reductase inhibitors. Trends Pharmacol. 1998;19:26–37.

76.

Davidson M, McKenney J, Stein E, Schrott H, Bakker-Arkema R, Fayyad R, Black D. Comparison of one-year efficacy and safety of atorvastatin versus lovastatin in primary hypercholesterolemia. Atorvastatin Study Group I. Am J Cardiol. 1997;79:1475–81.PubMed

77.

Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature. 1990;343:425–30.PubMed

78.

Graaf MR, Richel DJ, Van Noorden CJ, Guchelaar HJ. Effects of statins and farnesyltransferase inhibitors on the development and progression of cancer. Cancer Treat Rev. 2004;30:609–41.PubMed

79.

Casey PJ, Seabra MC. Protein prenyltransferases. J Biol Chem. 1996;271:5289–92.PubMed

80.

Auer J, Berent R, Weber T, Eber B. Clinical significance of pleiotropic effects of statins: lipid reduction and beyond. Curr Med Chem. 2002;9:1831–50.PubMed

81.

Menge T, Hartung HP, Stuve O. Statins – a cure-all for the brain? Nat Rev Neurosci. 2005;6:325–31.PubMed

82.

Esfandiari N, Khazaei M, Ai J, Bielecki R, Gotlieb L, Casper RF. Effect of a statin on an in vitro model of endometriosis. Fertil Steril. 2007;87:257–62.PubMed

83.

Cakmak H, Basar M, Seval-Celik Y, Osteen KG, Duleba AJ, Taylor HS, Lockwood CJ, Arici A. Statins inhibit monocyte chemotactic protein 1 expression in endometriosis. Reprod Sci. 2012;19:572–9.PubMedCentralPubMed

84.

Sharma I, Dhawan V, Mahajan N, Saha SC, Dhaliwal LK. In vitro effects of atorvastatin on lipopolysaccharide-induced gene expression in endometriotic stromal cells. Fertil Steril. 2010;94:1639–46.PubMed

85.

Oktem M, Esinler I, Eroglu D, Haberal N, Bayraktar N, Zeyneloglu HB. High-dose atorvastatin causes regression of endometriotic implants: a rat model. Hum Reprod. 2007;22:1474–80.PubMed

86.

Bruner-Tran KL, Osteen KG, Duleba AJ. Simvastatin protects against the development of endometriosis in a nude mouse model. J Clin Endocrinol Metab. 2009;94:2489–94.PubMedCentralPubMed

87.

Mraiche F, Cena J, Das D, Vollrath B. Effects of statins on vascular function of endothelin-1. Br J Pharmacol. 2005;144:715–26.PubMedCentralPubMed

88.

Ruperez M, Rodrigues-Diez R, Blanco-Colio LM, Sanchez-Lopez E, et al. HMG-CoA reductase inhibitors decrease angiotensin II-induced vascular fibrosis. Role of RhoA/ROCK and MAPK pathways. Hypertension. 2007;50:377–83.PubMed

89.

Hirsh J, Raschke R, Warrkentin TE, Dalen JE, Deykin D, Poller L. Heparin: mechanism of action, pharmacokinetics, dosing considerations, monitoring, efficacy, and safety. Chest. 1995;108:258–75.

90.

Becker RC. Optimizing heparin compounds: a working construct for future antithrombotic drug development. J Thromb Thrombolysis. 2004;18:55–8.PubMed

91.

Guidry C, Grinnell F. Heparin modulates the organization of hydrated collagen gels and inhibits gel contraction by fibroblasts. J Cell Biol. 1987;104:1097–103.PubMed

92.

Schaefer T, Roux M, Stuhlsatz HW, Herken R, Coulomb B, Krieg T, Smola H. Glycosaminoglycans modulate cell-matrix interactions of human fibroblasts and endothelial cells in vitro. J Cell Sci. 1996;109:479–88.PubMed

93.

Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape. Cell. 2007;128:635–8.PubMed

94.

Turner BM. Cellular memory and the histone code. Cell. 2002;111:285–91.PubMed

95.

Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33(Suppl):245–54.PubMed

96.

Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705.PubMed

97.

Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–97.PubMed

98.

Robertson KD, Wolffe AP. DNA methylation in health and disease. Nat Rev Genet. 2000;1:11–9.PubMed

99.

Rodenhiser D, Mann M. Epigenetics and human disease: translating basic biology into clinical applications. CMAJ. 2006;174:341–8.PubMedCentralPubMed

100.

Marks PA, Rifkind RA, Richon VM, Breslow R. Inhibitors of histone deacetylase are potentially effective anticancer agents. Clin Cancer Res. 2001;7:759–60.PubMed

101.

Norton VG, Imai BS, Yau P, Bradbury EM. Histone acetylation reduces nucleosome core particle linking number change. Cell. 1989;57:449–57.PubMed

102.

Choi CH, Burton ZF, Usheva A. Auto-acetylation of transcription factors as a control mechanism in gene expression. Cell Cycle. 2004;3:114–5.PubMed

103.

Joseph J, Mudduluru G, Antony S, Vashistha S, Ajitkumar P, Somasundaram K. Expression profiling of sodium butyrate (NaB)-treated cells: identification of regulation of genes related to cytokine signaling and cancer metastasis by NaB. Oncogene. 2004;23:6304–15.PubMed

104.

Wu Y, Strawn E, Basir Z, Halverson G, Guo SW. Promoter hypermethylation of progesterone receptor isoform B (PR-B) in endometriosis. Epigenetics. 2006;1:106–11.PubMed

105.

Wu Y, Strawn E, Basir Z, Halverson G, Guo SW. Aberrant expression of deoxyribonucleic acid methyltransferases DNMT1, DNMT3A, and DNMT3B in women with endometriosis. Fertil Steril. 2007;87:24–32.PubMed

106.

Xue Q, Lin Z, Yin P, Milad MP, Cheng Y-H, Confino E, Reierstad S, Bulun SE. Transcriptional activation of steroidogenic factor-1 by hypomethylation of the 5′ CpG island in endometriosis. J Clin Endocrinol Metab. 2007;92:3261–7.PubMed

107.

Xue Q, Lin Z, Cheng YH, Huang CC, Marsh E, Yin P, Milad MP, Confino E, Reierstad S, Innes J, Bulun SE. Promoter methylation regulates estrogen receptor 2 in human endometrium and endometriosis. Biol Reprod. 2007;77:681–7.PubMed

108.

Kawano Y, Nasu K, Li H, Tsuno A, Abe W, Takai N, Narahara H. Application of the histone deacetylase inhibitors for the treatment of endometriosis: histone modifications as pathogenesis and novel therapeutic target. Hum Reprod. 2011;26:2486–98.PubMed

109.

Nasu K, Kawano Y, Tsukamoto Y, Takano M, Takai N, Li H, Furukawa Y, Abe W, Moriyama M, Narahara H. Aberrant DNA methylation status of endometriosis: epigenetics as the pathogenesis, biomarker and therapeutic target. J Obstet Gynaecol Res. 2011;37:683–95.PubMed

110.

Abe W, Nasu K, Nakada C, Kawano Y, Moriyama M, Narahara H. miR-196b targets c-myc and Bcl-2 expression, inhibits proliferation and induces apoptosis in endometriotic stromal cells. Hum Reprod. 2013;28:750–61.PubMed

111.

Richon VM, O’Brien JP. Histone deacetylase inhibitors: a new class of potential therapeutic agents for cancer treatment. Clin Cancer Res. 2002;8:662–4.PubMed

112.

Verdin E, Dequiedt F, Kasler HG. Class II histone deacetylases: versatile regulators. Trends Genet. 2003;19:286–93.PubMed

113.

Cameron EE, Bachman KE, Myohanen S, Herman JG, Baylin SB. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet. 1999;21:103–7.PubMed

114.

Warrener R, Beamish H, Burgess A, Waterhouse NJ, Giles N, Fairlie D, Gabrielli B. Tumor cell-selective cytotoxicity by targeting cell cycle checkpoints. FASEB J. 2003;17:1550–2.PubMed

115.

Wu Y, Guo SW. Suppression of IL-1β-induced COX-2 expression by trichostatin A (TSA) in human endometrial stromal cells. Eur J Obstet Gynecol Reprod Biol. 2007;135:88–93.PubMed

116.

Wu Y, Starzinski-Powitz A, Guo SW. Trichostatin A, a histone deacetylase inhibitor, attenuates invasiveness and reactivates E-cadherin expression in immortalized endometriotic cells. Reprod Sci. 2007;14:374–82.PubMed

117.

Wu Y, Guo SW. Histone deacetylase inhibitors trichostatin A and valproic acid induce cell cycle arrest and p21 expression in immortalized human endometrial stromal cells. Eur J Obstet Gynecol Reprod Biol. 2008;137:198–203.PubMed

118.

Wu Y, Starzinski-Powitz A, Guo S-W. Prolonged stimulation with tumor necrosis factor-α induced partial methylation at PR-B promoter in immortalized epithelial-like endometriotic cells. Fertil Steril. 2008;90:234–7.PubMed

119.

Wu Y, Guo SW. Peroxisome proliferator-activated receptor-gamma and retinoid X receptor agonists synergistically suppress proliferation of immortalized endometrial stromal cells. Fertil Steril. 2009;91(Suppl):2142–7.PubMed

120.

Wu Y, Starzinski-Powitz A, Guo S-W. Constitutive and tumor necrosis factor-alpha-stimulated activation of nuclear factor-kappab in immortalized endometriotic cells and their suppression by trichostatin A. Gynecol Obstet Invest. 2010;70:23–33.PubMed

121.

Imesch P, Fink D, Fedier A. Romidepsin reduces histone deacetylase activity, induces acetylation of histones, inhibits proliferation, and activates apoptosis in immortalized epithelial endometriotic cells. Fertil Steril. 2010;94:2838–42.PubMed

122.

Imesch P, Samartzis EP, Schneider M, Fink D, Fedier A. Inhibition of transcription, expression, and secretion of the vascular epithelial growth factor in human epithelial endometriotic cells by romidepsin. Fertil Steril. 2011;95:1579–83.PubMed

123.

Lu Y, Nie J, Liu X, Zheng Y, Guo SW. Trichostatin A, a histone deacetylase inhibitor, reduces lesion growth and hyperalgesia in experimentally induced endometriosis in mice. Hum Reprod. 2010;25:1014–25.PubMed

124.

Liu M, Liu X, Zhang Y, Guo SW. Valproic acid and progestin inhibit lesion growth and reduce hyperalgesia in experimentally induced endometriosis in rats. Reprod Sci. 2012;19:360–73.PubMed

125.

Liu X, Guo SW. A pilot study on the off-label use of valproic acid to treat adenomyosis. Fertil Steril. 2008;89:246–50.PubMed



If you find an error or have any questions, please email us at admin@doctorlib.info. Thank you!