Semin Reprod Med 2020; 38(02/03): 168-178
DOI: 10.1055/s-0040-1718741
Review Article

Animal Models of Adenomyosis

Ryan M. Marquardt
1   Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, Michigan
2   Cell and Molecular Biology Program, Michigan State University, East Lansing, Michigan
,
1   Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, Michigan
,
Asgerally T. Fazleabas
1   Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, Michigan
› Author Affiliations
Funding Research reported in this publication was supported in part by the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health under award number R01HD084478 to J.W.J. and T32HD087166 to R.M.M, MSU AgBio Research, and Michigan State University. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Abstract

Adenomyosis is a nonmalignant uterine disorder in which endometrial tissue exists within and grows into the myometrium. Animal models have generated limited insight into the still-unclear pathogenesis of adenomyosis, provided a platform for preclinical screening of many drugs and compounds with potential as therapeutics, and elucidated mechanisms underlying the pain and fertility issues that occur in many women with the disease. Spontaneous adenomyosis has been studied in nonhuman primates, primarily in the form of case reports. Adenomyosis is routinely experimentally induced in mice through methods such as neonatal tamoxifen exposure, pituitary engraftment, and human tissue xenotransplantation. Several studies have also reported hormonal or environmental toxicant exposures that give rise to murine adenomyosis, and genetically engineered models have been created that recapitulate the human-like condition, most notably involving alteration of β-catenin expression. This review describes the animal models for adenomyosis and their contributions to our understanding of the factors underpinning the development of symptoms. Animal models represent a unique opportunity for understanding the molecular basis of adenomyosis and developing efficacious treatment options for affected women. Herein, we assess their different potentials and limitations with regard to identification of new therapeutic interventions and reflect on future directions for research and drug validation.



Publication History

Article published online:
26 October 2020

© 2020. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Yen CF, Huang SJ, Lee CL, Wang HS, Liao SK. Molecular characteristics of the endometrium in uterine adenomyosis and its biochemical microenvironment. Reprod Sci 2017; 24 (10) 1346-1361
  • 2 Aleksandrovych V, Basta P, Gil K. Current facts constituting an understanding of the nature of adenomyosis. Adv Clin Exp Med 2019; 28 (06) 839-846
  • 3 Levgur M. Therapeutic options for adenomyosis: a review. Arch Gynecol Obstet 2007; 276 (01) 1-15
  • 4 Koike N, Tsunemi T, Uekuri C. et al. Pathogenesis and malignant transformation of adenomyosis. (review) Oncol Rep 2013; 29 (03) 861-867
  • 5 Maier V, Holl M, Dietze R. et al. Adenomyotic glands are highly related to endometrial glands. Reprod Biomed Online 2020; 40 (06) 769-778
  • 6 Dior UP, Nisbet D, Fung JN. et al. The association of sonographic evidence of adenomyosis with severe endometriosis and gene expression in eutopic endometrium. J Minim Invasive Gynecol 2019; 26 (05) 941-948
  • 7 Hever A, Roth RB, Hevezi PA. et al. Molecular characterization of human adenomyosis. Mol Hum Reprod 2006; 12 (12) 737-748
  • 8 Habiba M. The animal model of adenomyosis. In: Uterine Adenomyosis. Springer; 2016: 123-127
  • 9 Greaves P, White IN. Experimental adenomyosis. Best Pract Res Clin Obstet Gynaecol 2006; 20 (04) 503-510
  • 10 Barrier BF, Allison J, Hubbard GB, Dick Jr EJ, Brasky KM, Schust DJ. Spontaneous adenomyosis in the chimpanzee (Pan troglodytes): a first report and review of the primate literature: case report. Hum Reprod 2007; 22 (06) 1714-1717
  • 11 Graham KJ, Hulst FA, Vogelnest L, Fraser IS, Shilton CM. Uterine adenomyosis in an orang-utan (Pongo abelii/pygmaeus). Aust Vet J 2009; 87 (01) 66-69
  • 12 Barrier BF, Malinowski MJ, Dick Jr EJ, Hubbard GB, Bates GW. Adenomyosis in the baboon is associated with primary infertility. Fertil Steril 2004; 82 (Suppl. 03) 1091-1094
  • 13 DiGiacomo RF. Gynecologic pathology in the rhesus monkey (Macaca mulatta). II. Findings in laboratory and free-ranging monkeys. Vet Pathol 1977; 14 (06) 539-546
  • 14 Baskin GB, Smith SM, Marx PA. Endometrial hyperplasia, polyps, and adenomyosis associated with unopposed estrogen in rhesus monkeys (Macaca mulatta). Vet Pathol 2002; 39 (05) 572-575
  • 15 Vannuccini S, Petraglia F. Recent advances in understanding and managing adenomyosis. F1000 Res 2019; 8: 8
  • 16 Carrarelli P, Yen CF, Funghi L. et al. Expression of inflammatory and neurogenic mediators in adenomyosis. Reprod Sci 2017; 24 (03) 369-375
  • 17 Vannuccini S, Tosti C, Carmona F. et al. Pathogenesis of adenomyosis: an update on molecular mechanisms. Reprod Biomed Online 2017; 35 (05) 592-601
  • 18 Braundmeier AG, Fazleabas AT. The non-human primate model of endometriosis: research and implications for fecundity. Mol Hum Reprod 2009; 15 (10) 577-586
  • 19 D'Hooghe TM, Kyama CM, Chai D. et al. Nonhuman primate models for translational research in endometriosis. Reprod Sci 2009; 16 (02) 152-161
  • 20 Fazleabas AT. Progesterone resistance in a baboon model of endometriosis. Semin Reprod Med 2010; 28 (01) 75-80
  • 21 Hastings JM, Fazleabas AT. A baboon model for endometriosis: implications for fertility. Reprod Biol Endocrinol 2006; 4 (Suppl. 01) S7
  • 22 Moses AS, Taratula OR, Lee H. et al. Nanoparticle-based platform for activatable fluorescence imaging and photothermal ablation of endometriosis. Small 2020; 16 (18) e1906936
  • 23 Slayden OD. Translational in vivo models for women's health: the nonhuman primate endometrium--a predictive model for assessing steroid receptor modulators. Handb Exp Pharmacol 2016; 232: 191-202
  • 24 Fazleabas AT, Brudney A, Gurates B, Chai D, Bulun S. A modified baboon model for endometriosis. Ann N Y Acad Sci 2002; 955: 308-317 , discussion 340–342, 396–406
  • 25 Waterton JC, Breen SA, Dukes M, Horrocks M, Wadsworth PF. A case of adenomyosis in a pigtailed monkey diagnosed by magnetic resonance imaging and treated with the novel pure antiestrogen, ICI 182,780. Lab Anim Sci 1993; 43 (03) 247-251
  • 26 Oh SJ, Shin JH, Kim TH. et al. β-Catenin activation contributes to the pathogenesis of adenomyosis through epithelial-mesenchymal transition. J Pathol 2013; 231 (02) 210-222
  • 27 Bellessort B, Bachelot A, Heude É. et al. Role of Foxl2 in uterine maturation and function. Hum Mol Genet 2015; 24 (11) 3092-3103
  • 28 Gonzalez G, Behringer RR. Dicer is required for female reproductive tract development and fertility in the mouse. Mol Reprod Dev 2009; 76 (07) 678-688
  • 29 Nagasawa H, Kusakawa S. Relationship between incidence and onset age of mammary tumours and uterine adenomyosis in four strains of mice: comparison with the findings of 40 generations previously. In Vivo 2001; 15 (04) 345-349
  • 30 Kida H. [Histological analysis of spontaneous adenomyosis-like changes in recombinant inbred mouse uterus (SMXA mouse)--a novel animal model for adenomyosis]. Nippon Sanka Fujinka Gakkai Zasshi 1994; 46 (04) 323-330
  • 31 Nagasawa H, Naito T. Enhanced potentials for mammary tumourigenesis and uterine adenomyosis in (SLN x C3H/He)F1 virgin mice. Lab Anim 1992; 26 (01) 23-24
  • 32 Nagasawa H, Mori T. Stimulation of mammary tumorigenesis and suppression of uterine adenomyosis by temporary inhibition of pituitary prolactin secretion during youth in mice (41492). Proc Soc Exp Biol Med 1982; 171 (02) 164-167
  • 33 Mori T, Nagasawa H, Takahashi S. The induction of adenomyosis in mice by intrauterine pituitary isografts. Life Sci 1981; 29 (12) 1277-1282
  • 34 Walker BE. Ovariectomy of adult mice exposed prenatally to diethylstilbestrol. Cancer Lett 1987; 34 (02) 115-120
  • 35 Koike E, Yasuda Y, Shiota M. et al. Exposure to ethinyl estradiol prenatally and/or after sexual maturity induces endometriotic and precancerous lesions in uteri and ovaries of mice. Congenit Anom (Kyoto) 2013; 53 (01) 9-17
  • 36 Parrott E, Butterworth M, Green A, White IN, Greaves P. Adenomyosis--a result of disordered stromal differentiation. Am J Pathol 2001; 159 (02) 623-630
  • 37 Tanwar PS, Lee HJ, Zhang L. et al. Constitutive activation of Beta-catenin in uterine stroma and smooth muscle leads to the development of mesenchymal tumors in mice. Biol Reprod 2009; 81 (03) 545-552
  • 38 Huang TS, Chen YJ, Chou TY. et al. Oestrogen-induced angiogenesis promotes adenomyosis by activating the Slug-VEGF axis in endometrial epithelial cells. J Cell Mol Med 2014; 18 (07) 1358-1371
  • 39 Suzuki-Kakisaka H, Murakami T, Hirano T, Terada Y, Yaegashi N, Okamura K. Selective accumulation of PpIX and photodynamic effect after aminolevulinic acid treatment of human adenomyosis xenografts in nude mice. Fertil Steril 2008; 90 (04) 1523-1527
  • 40 Chen YJ, Li HY, Huang CH. et al. Oestrogen-induced epithelial-mesenchymal transition of endometrial epithelial cells contributes to the development of adenomyosis. J Pathol 2010; 222 (03) 261-270
  • 41 Zhou S, Yi T, Liu R. et al. Proteomics identification of annexin A2 as a key mediator in the metastasis and proangiogenesis of endometrial cells in human adenomyosis. Mol Cell Proteomics 2012; 11: M112
  • 42 Koujyo T, Hatakeyama S, Yamada H. et al. Induction of endometriosis and adenomyosis by transvaginal pituitary transplantation in mice with and without natural killer cell activity. Am J Reprod Immunol 1998; 40 (06) 441-446
  • 43 Mori T, Ohta Y, Nagasawa H. Ultrastructural changes in uterine myometrium of mice with experimentally-induced adenomyosis. Experientia 1984; 40 (12) 1385-1387
  • 44 Mori T, Nagasawa H. Mechanisms of development of prolactin-induced adenomyosis in mice. Acta Anat (Basel) 1983; 116 (01) 46-54
  • 45 Matsuda M, Sasabe H, Adachi Y, Suzuki T, Mori T. Increased invasion activity of endometrial stromal cells and elevated expression of matrix metalloproteinase messenger RNA in the uterine tissues of mice with experimentally induced adenomyosis. Am J Obstet Gynecol 2001; 185 (06) 1374-1380
  • 46 Danilovich N, Roy I, Sairam MR. Emergence of uterine pathology during accelerated biological aging in FSH receptor-haploinsufficient mice. Endocrinology 2002; 143 (09) 3618-3627
  • 47 Yamashita M, Matsuda M, Mori T. Increased expression of prolactin receptor mRNA in adenomyotic uterus in mice. Life Sci 1997; 60 (17) 1437-1446
  • 48 Singtripop T, Mori T, Park MK, Sakamoto S, Kawashima S. Development of uterine adenomyosis after treatment with dopamine antagonists in mice. Life Sci 1991; 49 (03) 201-206
  • 49 Kelly MA, Rubinstein M, Asa SL. et al. Pituitary lactotroph hyperplasia and chronic hyperprolactinemia in dopamine D2 receptor-deficient mice. Neuron 1997; 19 (01) 103-113
  • 50 Huseby RA, Soares MJ, Talamantes F. Ectopic pituitary grafts in mice: hormone levels, effects on fertility, and the development of adenomyosis uteri, prolactinomas, and mammary carcinomas. Endocrinology 1985; 116 (04) 1440-1448
  • 51 Lipschutz A, Iglesias R, Panasevich VI, Salinas S. Pathological changes induced in the uterus of mice with the prolonged administration of progesterone and 19-nor-contraceptives. Br J Cancer 1967; 21 (01) 160-165
  • 52 Nagasawa H, Aoki M, Mori T. Effects of different schedules of progesterone treatment on mammary tumorigenesis and uterine adenomyosis in SHN virgin mice. Life Sci 1987; 40 (26) 2597-2602
  • 53 Nagasawa H, Aoki M, Sakagami N, Ishida M. Medroxyprogesterone acetate enhances spontaneous mammary tumorigenesis and uterine adenomyosis in mice. Breast Cancer Res Treat 1988; 12 (01) 59-66
  • 54 Ostrander PL, Mills KT, Bern HA. Long-term responses of the mouse uterus to neonatal diethylstilbestrol treatment and to later sex hormone exposure. J Natl Cancer Inst 1985; 74 (01) 121-135
  • 55 Bennett LM, McAllister KA, Malphurs J. et al. Mice heterozygous for a Brca1 or Brca2 mutation display distinct mammary gland and ovarian phenotypes in response to diethylstilbestrol. Cancer Res 2000; 60 (13) 3461-3469
  • 56 Walker BE. Tumors of female offspring of mice exposed prenatally to diethylstilbestrol. J Natl Cancer Inst 1984; 73 (01) 133-140
  • 57 Newbold RR, Jefferson WN, Padilla-Banks E. Long-term adverse effects of neonatal exposure to bisphenol A on the murine female reproductive tract. Reprod Toxicol 2007; 24 (02) 253-258
  • 58 Bruner-Tran KL, Duleba AJ, Taylor HS, Osteen KG. Developmental toxicant exposure is associated with transgenerational adenomyosis in a murine model. Biol Reprod 2016; 95 (04) 73
  • 59 Li Y, Zhang SF, Zou SE, Xia X, Bao L. Accumulation of nerve growth factor and its receptors in the uterus and dorsal root ganglia in a mouse model of adenomyosis. Reprod Biol Endocrinol 2011; 9: 30
  • 60 Mehasseb MK, Bell SC, Habiba MA. The effects of tamoxifen and estradiol on myometrial differentiation and organization during early uterine development in the CD1 mouse. Reproduction 2009; 138 (02) 341-350
  • 61 Mehasseb MK, Bell SC, Habiba MA. Neonatal administration of tamoxifen causes disruption of myometrial development but not adenomyosis in the C57/BL6J mouse. Reproduction 2010; 139 (06) 1067-1075
  • 62 Green AR, Styles JA, Parrott EL. et al. Neonatal tamoxifen treatment of mice leads to adenomyosis but not uterine cancer. Exp Toxicol Pathol 2005; 56 (4-5): 255-263
  • 63 Green AR, Edwards RE, Greaves P, White IN. Comparison of the effect of oestradiol, tamoxifen and raloxifene on nerve growth factor-alpha expression in specific neonatal mouse uterine cell types using laser capture microdissection. J Mol Endocrinol 2003; 30 (01) 1-11
  • 64 Acloque H, Adams MS, Fishwick K, Bronner-Fraser M, Nieto MA. Epithelial-mesenchymal transitions: the importance of changing cell state in development and disease. J Clin Invest 2009; 119 (06) 1438-1449
  • 65 Shen M, Liu X, Zhang H, Guo SW. Transforming growth factor β1 signaling coincides with epithelial-mesenchymal transition and fibroblast-to-myofibroblast transdifferentiation in the development of adenomyosis in mice. Hum Reprod 2016; 31 (02) 355-369
  • 66 Jin Z, Liu H, Xu C. Estrogen degrades Scribble in endometrial epithelial cells through E3 ubiquitin ligase HECW1 in the development of diffuse adenomyosis. Biol Reprod 2020; 102 (02) 376-387
  • 67 Schmalhofer O, Brabletz S, Brabletz T. E-cadherin, beta-catenin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev 2009; 28 (1-2): 151-166
  • 68 Arango NA, Szotek PP, Manganaro TF, Oliva E, Donahoe PK, Teixeira J. Conditional deletion of beta-catenin in the mesenchyme of the developing mouse uterus results in a switch to adipogenesis in the myometrium. Dev Biol 2005; 288 (01) 276-283
  • 69 Kawahara R, Matsuda M, Mori T. Increase in the number of integrinbeta1-immunoreactive monocyte-lineage cells in experimentally-induced adenomyosis in mice. Life Sci 2003; 73 (07) 907-916
  • 70 Marquardt RM, Kim TH, Shin JH, Jeong JW. Progesterone and estrogen signaling in the endometrium: What goes wrong in endometriosis?. Int J Mol Sci 2019; 20 (15) 20
  • 71 Che X, Wang J, He J, Guo X, Li T, Zhang X. The new application of mifepristone in the relief of adenomyosis-caused dysmenorrhea. Int J Med Sci 2020; 17 (02) 224-233
  • 72 Singtripop T, Mori T, Sakamoto S, Sassa S, Park MK, Kawashima S. Suppression of the development of uterine adenomyosis by danazol treatment in mice. Life Sci 1992; 51 (14) 1119-1125
  • 73 Otto C, Schkoldow J, Krahl E, Fuchs I, Ulbrich HF. Use of a murine endometriosis interna model for the characterization of compounds that effectively treat human endometriosis. Exp Ther Med 2012; 3 (03) 410-414
  • 74 Zhang X, Yuan H, Deng L, Hu F, Ma J, Lin J. Evaluation of the efficacy of a danazol-loaded intrauterine contraceptive device on adenomyosis in an ICR mouse model. Hum Reprod 2008; 23 (09) 2024-2030
  • 75 Selak V, Farquhar C, Prentice A, Singla A. Danazol for pelvic pain associated with endometriosis. Cochrane Database Syst Rev 2001; (04) CD000068
  • 76 Guo S, Lu X, Gu R, Zhang D, Sun Y, Feng Y. Transcriptome analysis of endometrial tissues following GnRH agonist treatment in a mouse adenomyosis model. Drug Des Devel Ther 2017; 11: 695-704
  • 77 Ferrero S, Evangelisti G, Barra F. Current and emerging treatment options for endometriosis. Expert Opin Pharmacother 2018; 19 (10) 1109-1125
  • 78 Zhou YF, Matsuda M, Mori T, Sakamoto S, Mitamura T. Effects of mifepristone (RU486) treatment on the development of uterine adenomyosis induced by pituitary grafting in mice. Life Sci 2000; 67 (22) 2713-2720
  • 79 Mori T, Kurata Y, Tabata Y, Niho N, Matsuda M, Zhou YF. Priming effects of novel nonsteroidal progesterone receptor modulators CP8816 and CP8863 on the development of adenomyosis in the mouse uterus. Life Sci 2002; 71 (05) 527-535
  • 80 Kurata Y, Tabata Y, Shinei R. et al. Endocrinological properties of two novel nonsteroidal progesterone receptor modulators, CP8816 and CP8863. J Pharmacol Exp Ther 2005; 313 (02) 916-920
  • 81 Nagasawa H, Noguchi Y, Mori T, Niki K, Namiki H. Suppression of normal and preneoplastic mammary growth and uterine adenomyosis with reduced growth hormone level in SHN mice given monosodium glutamate neonatally. Eur J Cancer Clin Oncol 1985; 21 (12) 1547-1551
  • 82 Mao X, Wang Y, Carter AV, Zhen X, Guo SW. The retardation of myometrial infiltration, reduction of uterine contractility, and alleviation of generalized hyperalgesia in mice with induced adenomyosis by levo-tetrahydropalmatine (l-THP) and andrographolide. Reprod Sci 2011; 18 (10) 1025-1037
  • 83 Chen Y, Zhu B, Zhang H, Liu X, Guo SW. Epigallocatechin-3-gallate reduces myometrial infiltration, uterine hyperactivity, and stress levels and alleviates generalized hyperalgesia in mice induced with adenomyosis. Reprod Sci 2013; 20 (12) 1478-1491
  • 84 Chen Y, Zhu B, Zhang H, Ding D, Liu X, Guo SW. Possible loss of GABAergic inhibition in mice with induced adenomyosis and treatment with epigallocatechin-3-gallate attenuates the loss with improved hyperalgesia. Reprod Sci 2014; 21 (07) 869-882
  • 85 Zhu B, Chen Y, Zhang H, Liu X, Guo SW. Resveratrol reduces myometrial infiltration, uterine hyperactivity, and stress levels and alleviates generalized hyperalgesia in mice with induced adenomyosis. Reprod Sci 2015; 22 (11) 1336-1349
  • 86 Nie J, Liu X. Leonurine attenuates hyperalgesia in mice with induced adenomyosis. Med Sci Monit 2017; 23: 1701-1706
  • 87 Feng T, Wei S, Wang Y. et al. Rhein ameliorates adenomyosis by inhibiting NF-κB and β-catenin signaling pathway. Biomed Pharmacother 2017; 94: 231-237
  • 88 Liu X, Guo SW. Valproic acid alleviates generalized hyperalgesia in mice with induced adenomyosis. J Obstet Gynaecol Res 2011; 37 (07) 696-708
  • 89 Jichan Nie, Xishi Liu, Guo SW. Promoter hypermethylation of progesterone receptor isoform B (PR-B) in adenomyosis and its rectification by a histone deacetylase inhibitor and a demethylation agent. Reprod Sci 2010; 17 (11) 995-1005
  • 90 Mori T, Yamasaki S, Masui F, Matsuda M, Sasabe H, Zhou YF. Suppression of the development of experimentally induced uterine adenomyosis by a novel matrix metalloproteinase inhibitor, ONO-4817, in mice. Exp Biol Med (Maywood) 2001; 226 (05) 429-433
  • 91 Zhou YF, Mori T, Kudo H, Asakai R, Sassa S, Sakamoto S. Effects of angiogenesis inhibitor TNP-470 on the development of uterine adenomyosis in mice. Fertil Steril 2003; 80 (Suppl. 02) 788-794
  • 92 Zhou YF, Mori T, Nagasawa H, Nakayama T, Kubota T, Sakamoto S. Probucol, a hypocholesterolemic agent, prevents the development of uterine adenomyosis induced by pituitary grafting in mice. Anticancer Res 2004; 24 (04) 2209-2212
  • 93 Zhu B, Chen Y, Shen X, Liu X, Guo SW. Anti-platelet therapy holds promises in treating adenomyosis: experimental evidence. Reprod Biol Endocrinol 2016; 14 (01) 66
  • 94 Sammour A, Pirwany I, Usubutun A, Arseneau J, Tulandi T. Correlations between extent and spread of adenomyosis and clinical symptoms. Gynecol Obstet Invest 2002; 54 (04) 213-216
  • 95 Guo S-W, Habiba M. Improving the preclinical mouse efficacy studies of adenomyosis. In: Uterine Adenomyosis. Springer; 2016: 129-139
  • 96 Harada T, Khine YM, Kaponis A, Nikellis T, Decavalas G, Taniguchi F. The impact of adenomyosis on women's fertility. Obstet Gynecol Surv 2016; 71 (09) 557-568
  • 97 Bruner-Tran KL, Osteen KG. Developmental exposure to TCDD reduces fertility and negatively affects pregnancy outcomes across multiple generations. Reprod Toxicol 2011; 31 (03) 344-350
  • 98 Mehasseb MK, Panchal R, Taylor AH. et al. Estrogen and progesterone receptor isoform distribution through the menstrual cycle in uteri with and without adenomyosis. Fertil Steril 2011; 95: 2228-2235
  • 99 Jeong JW, Lee HS, Franco HL. et al. beta-catenin mediates glandular formation and dysregulation of beta-catenin induces hyperplasia formation in the murine uterus. Oncogene 2009; 28 (01) 31-40
  • 100 Guo S, Li Z, Yan L, Sun Y, Feng Y. GnRH agonist improves pregnancy outcome in mice with induced adenomyosis by restoring endometrial receptivity. Drug Des Devel Ther 2018; 12: 1621-1631
  • 101 McEntee K, Nielsen SW. Tumours of the female genital tract. Bull World Health Organ 1976; 53 (2-3): 217-226
  • 102 Stöcklin-Gautschi NM, Guscetti F, Reichler IM, Geissbühler U, Braun SA, Arnold S. Identification of focal adenomyosis as a uterine lesion in two dogs. J Small Anim Pract 2001; 42 (08) 413-416
  • 103 Gelberg HB, McEntee K. Pathology of the canine and feline uterine tube. Vet Pathol 1986; 23 (06) 770-775
  • 104 Baldi A, Lanza A, Menicagli F, Signorile PG, Spugnini EP. Histological and immunohistochemical characterization of a case of endometriosis in a guinea pig (Cavia tschudii) . Case Rep Vet Med 2017; 2017: 4594510
  • 105 Meissner WA, Sommers SC, Sherman G. Endometrial hyperplasia, endometrial carcinoma, and endometriosis produced experimentally by estrogen. Cancer 1957; 10 (03) 500-509
  • 106 Mori T, Kyokuwa M, Nagasawa H. Animal model of uterine adenomyosis: induction of the lesion in rats by ectopic pituitary isografting. Lab Anim Sci 1998; 48 (01) 64-68
  • 107 Sengupta P, Sharma A, Mazumdar G. et al. The possible role of fluoxetine in adenomyosis: an animal experiment with clinical correlations. J Clin Diagn Res 2013; 7 (07) 1530-1534