Profile of Selected Cytokines and Growth Factors in Uterine Myomas in Females of Various Age

Authors

  • P. Madej Department of Endocrinological Gynecology, Medical University of Silesia, Katowice, Poland
  • D. Plewka Department of Histology and Embryology, Medical University of Silesia, Katowice, Poland
  • D. Pluta Department of Endocrinological Gynecology, Medical University of Silesia, Katowice, Poland
  • R. Bilski Department of Endocrinological Gynecology, Medical University of Silesia, Katowice, Poland
  • G. Franik Department of Endocrinological Gynecology, Medical University of Silesia, Katowice, Poland
  • A. Madej-Czech Department of Intensive Care and Neonatal Pathology Medical University of Silesia, Zabrze, Poland
  • M. Maksym Department of Endocrinological Gynecology, Medical University of Silesia, Katowice, Poland
  • M. Nylec Department of Endocrinological Gynecology, Medical University of Silesia, Katowice, Poland
  • S. Czech District Hospital of Orthopedics and Trauma Surgery, Piekary lskie, Poland

DOI:

https://doi.org/10.14205/2309-4400.2014.02.02.2

Keywords:

Uterine myomas, interleukins, tumour necrosis factor, transforming growth factors, insulin-like growth factors, age.

Abstract

 Objective: Uterine myomas are the most common non-malignant tumours in adult females. Causes of development and progression of the tumour remain unclear but, apart from steroid hormones, factors which stimulate growth of uterine myomas include cytokines and growth factors. In this study an analysis was conducted of cytokine environment in myomas of various size and in healthy uterine tissue in women of various age. Methods: Concentrations of selected cytokines and growth factors were evaluated using immunohistochemical techniques in small (<3cm) and large (>5cm) uterine myomas, in women of generative age or perimenopausal age. Immunohistochemical studies were performed on properly prepared paraffin sections, to which the primary antibodies was applied. The obtained immunohistochemical reactions were evaluated under a light microscope. The results were subjected to statistical analysis. Photographic documentation was prepared with photographic attachment. Results: Interleukin-1 level in young women in small myomas was higher than in the control, amounting to 345%. In large myomas - it was 415%. In women of perimenopausal age in small myomas was augmented to 320% and in large myomas - 240%. Interleukin-6 level in young women in small myomas was higher than in the control, reaching 255%. In large myomas - it was 285%. In women of perimenopausal age in small myomas was augmented to 240% and in large myomas - 235%. TNF-a level in young women in small myomas was higher than that in the control, amounting to 265%. In large myomas - it was 230%. In women of perimenopausal age in small myomas was augmented to 270% and in large myomas - 140%. TGF-β1, TGF-β2, TGF-β3 levels in the young women in small myomas amounting to 310%, 285%, 335% of the control levels for, respectively, TGF-β1, TGF-β2 and TGF-β3. In large myomas - it was respectively 390%, 375%, 320%. In women of perimenopausal age in small myomas was augmented, respectively, to 260%, 270%, 385% and in large myomas - 295%, 295%, 355%. IGF-1 and IGF-2 levels in young women in small myomas was higher than in the control, amounting to respectively 210% and 250%. In large myomas - it was respectively 290% and 200%. In women of perimenopausal age in small myomas was augmented, respectively, to 245% and 195% and in large myomas - 265% and 215%. Conclusions: in myomatous uterine cells of young women an increase was noted in expression of IL-1 and IL-6 both in small and in large myomas. In women of perimenopausal age, the increase in expression of TNF-α took place only in small myomas with lower expression in large myomas. In women of generative age subjected to evaluation of transforming growth factors, high expression of the peptide was detected in all groups of leiomyomas. In both types of leiomyomas a comparable expression of insulin-like growth factors was detected, irrespectively of women's age.

References

Flake GP, Andersen J, Dixon D. Etiology and pathogenesis of uterine leyomyomas: a review. Environ Helth Persp 2003; 111: 1037-54. http://dx.doi.org/10.1289/ehp.5787

Robak-Choubek D, Jakiel G. Uterine myomas (in Polish). Przegl Menopauzalny 2006; 6: 409 12.

Parker WH. Etiology, symptomatology, and diagnosis of uterine myomas. Fertil Steril 2007; 87: 725-36. http://dx.doi.org/10.1016/j.fertnstert.2007.01.093

Andersen J, Barbieri RL. Abnormal gene expression in uterine leiomyomas. J Soc Gynecol Invest 1995; 2: 663-72. http://dx.doi.org/10.1016/1071-5576(95)00021-6

Walker CL, Stewart EA. Uterine fibroids: the elephant in the room. Science 2005; 308: 1589-92. http://dx.doi.org/10.1126/science.1112063

Otubu JA, Buttram VC, Besch NF, Besch PK. Unconjugated steroids in leiomyomas and tumor-bearing myometrium. Am J Obstet Gynecol 1982; 143: 130-133.

Sozen I, Arici A. Interactions of cytokines, growth factors, and the extracellular matrix in the cellular biology of uterine leiomyomata. Fertil Steril 2002; 78: 1-12. http://dx.doi.org/10.1016/S0015-0282(02)03154-0

Joseph DS, Malik M, Nurudeen S, Catherino WH. Myometrial cells undergo fibrotic transformation under the influence of transforming growth factor beta-3. Fertil Steril 2010; 93: 1500-8. http://dx.doi.org/10.1016/j.fertnstert.2009.01.081

Arici A, Sozen I. Transforming growth factor-b3 is expressed at high levels in leiomyoma where it stimulates fibronectin expression and cell proliferation. Fertil Steril 2000; 73: 1006- 11. http://dx.doi.org/10.1016/S0015-0282(00)00418-0

Manni A, Wei L, Badger B, et al. Expression of messenger RNA for insulin-like growth factors and insulin-like growth factor binding proteins by experimental breast cancer and normal breast tissue in vivo. Endocrinology 1992; 130: 1744- 6.

Chandrasekhar Y, Heiner J, Osuamkpe C, Nagamani M. Insulin-like growth factor I and II binding in human myometrium and leiomyomas. Am J Obstet Gynecol 1992; 166: 64-9. http://dx.doi.org/10.1016/0002-9378(92)91831-T

Wola ska M, Ba kowski E. An accumulation of insulin-like growth factor I (IGF-I) in human myometrium and uterine leiomyomas in various stages of tumour growth. Eur Cytokine Net 2004; 15: 359-63.

Burroughs KD, Howe SR, Okubo Y, Fuchs-Young R, LeRoith D, Walker CL. Dysregulation of IGF-I signaling in uterine leiomyoma. J Endocrinol 2002; 172: 83-93. http://dx.doi.org/10.1677/joe.0.1720083

Iwabe T, Harada T, Tsudo T, Tanikawa M, Onohara Y, Terakawa N. Pathogenetic significance of increased levels of interleukin-8 in the peritoneal fluid of patients with endometriosis. Fertil Steril 1998; 69: 924-30. http://dx.doi.org/10.1016/S0015-0282(98)00049-1

Rier SE, Zarmakoupis PN, Hu X, Becker JL. Dysregulation of interleukin-6 responses in ectopic endometrial stromal cells: correlation with decreased soluble receptor levels in peritoneal fluid of women with endometriosis. J Clin Endocrinol Metab 1995; 80: 1431-7.

Tsudo T, Harada T, Iwabe T, et al. Altered gene expression and secretion of interleukin-6 in stromal cells derived from endometriotic tissues. Fertil Steril 2000; 73: 205-11. http://dx.doi.org/10.1016/S0015-0282(99)00496-3

Sakamoto Y, Harada T, Horie S, Iba Y, Taniguchi F, Yoshida S. Tumor necrosis factor- -induced interleukin-8 (IL-8) expression in endometriotic stromal cells, probably through nuclear factor-B activation: gonadotropin-releasing hormone agonist treatment reduced IL-8 expression. J Clin Endocrinol Metab 2003; 88: 730-5. http://dx.doi.org/10.1210/jc.2002-020666

Yamauchi N, Harada T, Taniguchi F, Yoshida S, Iwabe T, Terakawa N. Tumor necrosis factor- induced the release of interleukin-6 from endometriotic stromal cells by the nuclear factor-B and mitogen activated protein kinase pathways. Fertil Steril 2004; 82(Suppl 3): 1023-8. http://dx.doi.org/10.1016/j.fertnstert.2004.02.134

Wang Y, Yang J, Gao Y, et al. Regulatory effect of E2, IL 6 and IL-8 on the growth of epithelial ovarian cancer cells. Cell Mol Immunol 2005; 2: 365-72.

Palena C, Hamilton DH, Fernando RI. Influence of IL-8 on the epithelial-mesenchymal transition and the tumor microenvironment Future Oncol 2012; 8: 713-22. http://dx.doi.org/10.2217/fon.12.59

ukaszewicz M, Mroczko B, Szmitkowski M. Clinical significance of interleukin 6 (IL-6) as a prognostic factor in a neoplastic disease (in Polish). Pol Arch Med Wewn 2007; 117: 247-51.

Roy D, Sarkar S, Felty Q. Levels of IL-1 control stimulatory/inhibitory growth of cancer cells. Front Biosci 2006; 11: 889-98. http://dx.doi.org/10.2741/1845

Tsudo T, Harada T, Iwabe T, et al. Altered gene expression and secretion of interleukin-6 in stromal cells derived from endometriotic tissues. Fertil Steril 2000; 73: 205-11. http://dx.doi.org/10.1016/S0015-0282(99)00496-3

Tagashira Y, Taniguchi F, Harada T, Ikeda A, Watanabe A, Terakawa N. Interleukin-10 attenuates TNF-alpha-induced interleukin-6 production in endometriotic stromal cells. Fertil Steril 2009; 91(5 Suppl): 2185-92. http://dx.doi.org/10.1016/j.fertnstert.2008.04.052

Kulbe H, Thompson R, Wilson JL, et al. The inflammatory cytokine tumor necrosis factor- generates an autocrine tumor-promoting network in epithelial ovarian cancer cells. Cancer Res 2007; 67: 585-92. http://dx.doi.org/10.1158/0008-5472.CAN-06-2941

Szlosarek PW, Grimshaw MJ, Kulbe H, et al. Expression and regulation of tumor necrosis factor alpha in normal and malignant ovarian epithelium. Mol Cancer Ther 2006; 5: 382- 90. http://dx.doi.org/10.1158/1535-7163.MCT-05-0303

Janes KA, Gaudet S, Albeck JG, Nielsen UB, Lauffenburger DA, Sorger PK. The response of human epithelial cells to TNF involves an inducible autocrine cascade. Cell 2006; 124: 1225-39. http://dx.doi.org/10.1016/j.cell.2006.01.041

Madhusudan S, Foster M, Muthuramalingam SR, et al. A phase II study of ethanercept (ENBREL) a tumour necrosis factor-a inhibitor in recurrent ovarian cancer. J Clin Oncol 2005; 10: 6528–34.

Zhu L, Skoultchi AI. Coordinating cell proliferation and differentiation. Curr Opin Genet Dev 2001; 11: 91-7. http://dx.doi.org/10.1016/S0959-437X(00)00162-3

McClellan KA, Slack RS. Novel functions for cell cycle genes in nervous system development. Cell Cycle 2006; 5: 1506- 13. http://dx.doi.org/10.4161/cc.5.14.2980

Arici A, Sozen I. Expression, menstrual cycle-dependent activation, and bimodal mitogenic effect of transforming growth factor-beta1 in human myometrium and leiomyoma. Am J Obstet Gynecol 2003; 188: 76-83. http://dx.doi.org/10.1067/mob.2003.118

Zeyneloglu HB, Esinler I, Ozdemir BH, Haydardedeoglu B, Oktem M, Batioglu S. Immunohistochemical characteristics of intramural leiomyomata that enlarge during controlled ovarian hyperstimulation for in vitro fertilization. Gynecol Obstet Invest 2008; 65: 252-7. http://dx.doi.org/10.1159/000113049

Wight TN. Versican: a versatile extracellular matrix proteoglycan in cell biology. Curr Opin Cell Biol 2002; 14: 617-23. http://dx.doi.org/10.1016/S0955-0674(02)00375-7

Rogers R, Norian J, Malik M, et al. Mechanical homeostasis is altered in uterine leiomyoma. Am J Obstet Gynecol 2008; 198: 474.e1-11.

Mitropoulou TN, Theocharis AD, Stagiannis KD, Karamanos NK. Identification, quantification and fine structural characterization of glycosaminoglycans from uterine leiomyoma and normal myometrium. Biochimie 2001; 83: 529-36. http://dx.doi.org/10.1016/S0300-9084(01)01281-0

Berto AG, Sampaio LO, Franco CR, Cesar RM Jr, Michelacci YM. A comparativeanalysis of structure and spatial distribution of decorin in human leiomyoma and normal myometrium. Biochim Biophys Acta 2003; 1619: 98-112. http://dx.doi.org/10.1016/S0304-4165(02)00446-4

Bierie B, Moses HL. Tumour microenvironment: TGF-: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer 2006; 6: 506-20. http://dx.doi.org/10.1038/nrc1926

Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor in human disease. N Engl J Med 2000; 342: 1350-8. http://dx.doi.org/10.1056/NEJM200005043421807

He W, Dorn DC, Erdjument-Bromage H, Tempst P, Moore MA, Massague J. Hematopoiesis controlled by distinct TIF1gamma and Smad4 branches of the TGF- pathway. Cell 2006; 125: 929-41. http://dx.doi.org/10.1016/j.cell.2006.03.045

Moustakas A, Souchelnytskyi S, Heldin CH. Smad regulation in TGF- signal transduction. J Cell Sci 2001; 114: 4359-69.

Dijke P, Hill CS. New insights into TGF--Smad signaling. Trends Biochem Sci 2004; 29: 265-73. http://dx.doi.org/10.1016/j.tibs.2004.03.008

Joseph DS, Malik M, Nurudeen S, Catherino WH. Myometrial cells undergo fibrotic transformation under the influence of transforming growth factor beta-3. Fertil Steril 2010; 93: 1500-8. http://dx.doi.org/10.1016/j.fertnstert.2009.01.081

Leppert PC, Catherino WH, Segars JH. A new hypothesis about the origin of uterine fibroids based on gene expression profiling with microarrays. Am J Obstet Gynecol 2006; 195: 415-20. http://dx.doi.org/10.1016/j.ajog.2005.12.059

Abreu JG, Ketpura NI, Reversade B, Robertis EM. Connective-tissue growth factor (CTGF) modulates cell signaling by BMP and TGF-. Nat Cell Biol 2002; 4: 599-604.

Muroski ME, Roycik MD, Newcomer RG, Van den Steen PE, Opdenakker G, Monroe HR. Matrix metalloproteinase9/gelatinase B is a putative therapeutic target of chronic obstructive pulmonary disease and multiple sclerosis. Curr Pharm Biotechnol 2008; 9: 34-46. http://dx.doi.org/10.2174/138920108783497631

Norian JM, Malik M, Parker CY, et al. Transforming growth factor 3 regulates the versican variants in the extracellular matrix-rich uterine leiomyomas. Reprod Sci 2009; 16: 1153- 64. http://dx.doi.org/10.1177/1933719109343310

Leppert PC, Baginski T, Prupas C, Catherino WH, Pletcher S, Segars JH. Comparative ultrastructure of collagen fibrils in uterine leiomyomas and normal myometrium. Fertil Steril 2004; 82: 1182-7. http://dx.doi.org/10.1016/j.fertnstert.2004.04.030

Malik M, Catherino WH. Novel method to characterize primary cultures of leiomyoma and myometrium with the use of confirmatory biomarker gene arrays. Fertil Steril 2007; 87: 1166-72. http://dx.doi.org/10.1016/j.fertnstert.2006.08.111

Ding L, Xu J, Luo X, Chegini N. Gonadotropin releasing hormone and transforming growth factor beta activate mitogen-activated protein kinase/extracellularly regulated kinase and differentially regulate fibronectin, type I collagen, and plasminogen activator inhibitor-1 expression in leiomyoma and myometrial smooth muscle cells. J Clin Endocrinol Metab 2004; 89: 5549-57. http://dx.doi.org/10.1210/jc.2004-0161

Wang W, Schulze CJ, Suarez-Pinzon WL, Dyck JR, Sawicki G, Schulz R. Intracellular action of matrix metalloproteinase-2 accounts for acute myocardial ischemia and reperfusion injury. Circulation 2002; 106: 1543-9. http://dx.doi.org/10.1161/01.CIR.0000028818.33488.7B

Sawicki G, Leon H, Sawicka J, et al. Degradation of myosin light chain in isolated rat hearts subjected to ischemiareperfusion injury: a new intracellular target for matrix metalloproteinase-2. Circulation 2005; 112: 544-52. http://dx.doi.org/10.1161/CIRCULATIONAHA.104.531616

Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res 2006; 69: 562-73. http://dx.doi.org/10.1016/j.cardiores.2005.12.002

Préaux AM, Mallat A, Nhieu JT, D'Ortho MP, Hembry RM, Mavier P. Matrix metalloproteinase-2 activation in human hepatic fibrosis regulation by cell-matrix interactions. Hepatology 1999; 30: 944-50. http://dx.doi.org/10.1002/hep.510300432

Bogusiewicz M, Stryjecka-Zimmer M, Postawski K, Jakimiuk AJ, Rechberger T. Activity of matrix metalloproteinase-2 and -9 and contents of their tissue inhibitors in uterine leiomyoma and corresponding myometrium. Gynecol Endocrinol 2007; 23: 541-6. http://dx.doi.org/10.1080/09513590701557416

Lee BS, Nowak RA. Human leiomyoma smooth muscle cells show increased expression of transforming growth factorbeta 3 (TGF beta 3) and altered responses to the antiproliferative effects of TGF beta. J Clin Endocrinol Metab 2001; 86: 913-20.

Chegini N, Ma C, Tang XM, Williams RS. Effects of GnRH analogues, “add-back” steroid therapy, antiestrogen and antiprogestins on leiomyoma and myometrial smooth muscle cell growth and transforming growth factor-beta expression. Mol Hum Reprod 2002; 8: 1071-8. http://dx.doi.org/10.1093/molehr/8.12.1071

Dou Q, Zhao Y, Tarnuzzer RW, et al. Suppression of transforming growth factor- (TGF- ) and TGF- receptor messenger ribonucleic acid and protein expression in leiomyomata in women receiving gonadotropin-releasing hormone agonist therapy. J Clin Endocrinol Metab 1996; 81: 3222-30.

De Falco M, Staibano S, D'Armiento FP, et al. Preoperative treatment of uterine leiomyomas: clinical findings and expression of transforming growth factor-beta3 and connective tissue growth factor. J Soc Gynecol Investig 2006; 13: 297-303. http://dx.doi.org/10.1016/j.jsgi.2006.02.008

Kunz-Schughart LA, Knuechel R. Tumor-associated fibroblasts (part I): Active stromal participants in tumor development and progression? Histol Histopathol 2002; 17: 599-621.

Chechliska M. Role of cytokines in neoplastic processes (in Polish). Nowotwory. Journal of Oncology 2003; 53: 648-59.

Maruo T, Ohara N, Wang J, Matsuo H. Sex steroidal regulation of uterine leiomyoma growth and apoptosis. Hum Reprod Update 2004; 10: 207-220. http://dx.doi.org/10.1093/humupd/dmh019

Gao Z, Matsuo H, Wang Y, Nakago S, Maruo T. Upregulation by IGF-I of proliferating cell nuclear antigen and Bcl-2 protein expression in human uterine leiomyoma cells. J Clin Endocrinol Metab 2001; 86: 5593-9. http://dx.doi.org/10.1210/jcem.86.11.8008

Van der Ven LTM, Roholl PJM, Gloudemans T, et al. Expression of insulin-like growth factors (IGFs), their receptors and IGF binding protein-3 in normal, benign and malignant smooth muscle tissues. Br J Cancer 1997; 75: 1631-40. http://dx.doi.org/10.1038/bjc.1997.278

Englund K, Lindblom B, Carlström K, Gustavsson I, Sjöblom P, Blanck A. Gene expression and tissue concentrations of IGF-I in human miometrium and fibroids under different hormonal conditions. Mol Hum Reprod 2000; 6: 915-20. http://dx.doi.org/10.1093/molehr/6.10.915

Dixon D, He H, Haseman JK. Immunohistological localization of growth factors and their receptors in uterine leiomyomas and matched myometrium. Environ Health Perspect 2000; 108 (Suppl. 5): 795-802. http://dx.doi.org/10.1289/ehp.00108s5795

Schubert G, Elger W, Kaufmann G, Schneider B, Reddersen G, Chwalisz K. Discovery, chemistry, and reproductive pharmacology of asoprisnil and related 11 -benzaldoxime substituted selective progesterone receptor modulators (SPRMs). Semin Reprod Med 2005; 23: 58-73. http://dx.doi.org/10.1055/s-2005-864034

Downloads

Published

2014-06-30

Issue

Section

Articles