#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

mTOR Directs Breast Morphogenesis through the PKC-alpha-Rac1 Signaling Axis


The protein kinase mTOR is frequently activated in breast cancers, where it enhances cancer cell growth, survival, and metastastic spread to distant organs. Thus, mTOR is an attractive, clinically relevant molecular target for drugs designed to treat metastatic breast cancers. However, mTOR exists in two distinct complexes, mTORC1 and mTORC2, and the relative roles of each complex have not been elucidated. Moreover, as pathways that regulate normal tissue growth and development are often highjacked to promote cancer, understanding mTOR function in normal mammary epithelial development will likely provide insight into its role in tumor progression. In this study, we assessed the role of mTORC1 and mTORC2 complexes in normal mammary epithelial cell branching, survival, and invasion. Interestingly, while mTORC1 was not required for branching, survival and invasion of mammary epithelial cells, mTORC2 was necessary for these processes in both mouse and human models. Furthermore, we found that mTORC2 exerts its effects primarily through downstream activation of a PKC-alpha-Rac1 signaling axis rather than the more well-studied Akt signaling pathway. Our studies identify a novel role for the mTORC2 complex in mammary morphogenesis, including cell survival and motility, which are relevant to breast cancer progression.


Vyšlo v časopise: mTOR Directs Breast Morphogenesis through the PKC-alpha-Rac1 Signaling Axis. PLoS Genet 11(7): e32767. doi:10.1371/journal.pgen.1005291
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005291

Souhrn

The protein kinase mTOR is frequently activated in breast cancers, where it enhances cancer cell growth, survival, and metastastic spread to distant organs. Thus, mTOR is an attractive, clinically relevant molecular target for drugs designed to treat metastatic breast cancers. However, mTOR exists in two distinct complexes, mTORC1 and mTORC2, and the relative roles of each complex have not been elucidated. Moreover, as pathways that regulate normal tissue growth and development are often highjacked to promote cancer, understanding mTOR function in normal mammary epithelial development will likely provide insight into its role in tumor progression. In this study, we assessed the role of mTORC1 and mTORC2 complexes in normal mammary epithelial cell branching, survival, and invasion. Interestingly, while mTORC1 was not required for branching, survival and invasion of mammary epithelial cells, mTORC2 was necessary for these processes in both mouse and human models. Furthermore, we found that mTORC2 exerts its effects primarily through downstream activation of a PKC-alpha-Rac1 signaling axis rather than the more well-studied Akt signaling pathway. Our studies identify a novel role for the mTORC2 complex in mammary morphogenesis, including cell survival and motility, which are relevant to breast cancer progression.


Zdroje

1. Gjorevski N. and Nelson C.M., Integrated morphodynamic signalling of the mammary gland. Nat Rev Mol Cell Biol, 2011. 12(9): p. 581–93. doi: 10.1038/nrm3168 21829222

2. McNally S. and Martin F., Molecular regulators of pubertal mammary gland development. Ann Med, 2011. 43(3): p. 212–34. doi: 10.3109/07853890.2011.554425 21417804

3. Laplante M. and Sabatini D.M., mTOR signaling in growth control and disease. Cell, 2012. 149(2): p. 274–93. doi: 10.1016/j.cell.2012.03.017 22500797

4. Phung T.L., et al., Pathological angiogenesis is induced by sustained Akt signaling and inhibited by rapamycin. Cancer Cell, 2006. 10(2): p. 159–70. 16904613

5. Sarbassov D.D., et al., Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell, 2006. 22(2): p. 159–68. 16603397

6. Rosner M. and Hengstschlager M., Cytoplasmic and nuclear distribution of the protein complexes mTORC1 and mTORC2: rapamycin triggers dephosphorylation and delocalization of the mTORC2 components rictor and sin1. Hum Mol Genet, 2008. 17(19): p. 2934–48. doi: 10.1093/hmg/ddn192 18614546

7. Hong, S.M., et al., Rapamycin inhibits both motility through down-regulation of p-STAT3 (S727) by disrupting the mTORC2 assembly and peritoneal dissemination in sarcomatoid cholangiocarcinoma. Clin Exp Metastasis, 2012.

8. Jankiewicz M., Groner B., and Desrivieres S., Mammalian target of rapamycin regulates the growth of mammary epithelial cells through the inhibitor of deoxyribonucleic acid binding Id1 and their functional differentiation through Id2. Mol Endocrinol, 2006. 20(10): p. 2369–81. 16772532

9. Kim S.H., Zukowski K., and Novak R.F., Rapamycin effects on mTOR signaling in benign, premalignant and malignant human breast epithelial cells. Anticancer Res, 2009. 29(4): p. 1143–50. 19414357

10. Galbaugh T., et al., EGF-induced activation of Akt results in mTOR-dependent p70S6 kinase phosphorylation and inhibition of HC11 cell lactogenic differentiation. BMC Cell Biol, 2006. 7: p. 34. 16984645

11. Moriya H., et al., Secretion of three enzymes for fatty acid synthesis into mouse milk in association with fat globules, and rapid decrease of the secreted enzymes by treatment with rapamycin. Arch Biochem Biophys, 2011. 508(1): p. 87–92. doi: 10.1016/j.abb.2011.01.015 21281598

12. Pauloin A. and Chanat E., Prolactin and epidermal growth factor stimulate adipophilin synthesis in HC11 mouse mammary epithelial cells via the PI3-kinase/Akt/mTOR pathway. Biochim Biophys Acta, 2012. 1823(5): p. 987–96. doi: 10.1016/j.bbamcr.2012.02.016 22426621

13. Andrechek E.R., et al., Amplification of the neu/erbB-2 oncogene in a mouse model of mammary tumorigenesis. Proc Natl Acad Sci U S A, 2000. 97(7): p. 3444–9. 10716706

14. Shiota C., et al., Multiallelic disruption of the rictor gene in mice reveals that mTOR complex 2 is essential for fetal growth and viability. Dev Cell, 2006. 11(4): p. 583–9. 16962829

15. Burnett P.E., et al., RAFT1 phosphorylation of the translational regulators p70 S6 kinase and 4E-BP1. Proc Natl Acad Sci U S A, 1998. 95(4): p. 1432–7. 9465032

16. Ewald A.J., et al., Collective epithelial migration and cell rearrangements drive mammary branching morphogenesis. Dev Cell, 2008. 14(4): p. 570–81. doi: 10.1016/j.devcel.2008.03.003 18410732

17. McDonald P.C., et al., Rictor and integrin-linked kinase interact and regulate Akt phosphorylation and cancer cell survival. Cancer Res, 2008. 68(6): p. 1618–24. doi: 10.1158/0008-5472.CAN-07-5869 18339839

18. Zhang F., et al., mTOR complex component Rictor interacts with PKCzeta and regulates cancer cell metastasis. Cancer Res, 2010. 70(22): p. 9360–70. doi: 10.1158/0008-5472.CAN-10-0207 20978191

19. Agarwal N.K., et al., Rictor regulates cell migration by suppressing RhoGDI2. Oncogene, 2013. 32(20): p. 2521–6. doi: 10.1038/onc.2012.287 22777355

20. Schwertfeger K.L., Richert M.M., and Anderson S.M., Mammary gland involution is delayed by activated Akt in transgenic mice. Mol Endocrinol, 2001. 15(6): p. 867–81. 11376107

21. Ikenoue T., et al., Essential function of TORC2 in PKC and Akt turn motif phosphorylation, maturation and signalling. EMBO J, 2008. 27(14): p. 1919–31. doi: 10.1038/emboj.2008.119 18566587

22. Wullschleger S., Loewith R., and Hall M.N., TOR signaling in growth and metabolism. Cell, 2006. 124(3): p. 471–84. 16469695

23. Iden S. and Collard J.G., Crosstalk between small GTPases and polarity proteins in cell polarization. Nat Rev Mol Cell Biol, 2008. 9(11): p. 846–59. doi: 10.1038/nrm2521 18946474

24. Muthuswamy S.K. and Xue B., Cell polarity as a regulator of cancer cell behavior plasticity. Annu Rev Cell Dev Biol, 2012. 28: p. 599–625. doi: 10.1146/annurev-cellbio-092910-154244 22881459

25. Sun S.Y., et al., Activation of Akt and eIF4E survival pathways by rapamycin-mediated mammalian target of rapamycin inhibition. Cancer Res, 2005. 65(16): p. 7052–8. 16103051

26. O'Reilly K.E., et al., mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res, 2006. 66(3): p. 1500–8. 16452206

27. Wang X., et al., Enhancing mammalian target of rapamycin (mTOR)-targeted cancer therapy by preventing mTOR/raptor inhibition-initiated, mTOR/rictor-independent Akt activation. Cancer Res, 2008. 68(18): p. 7409–18. doi: 10.1158/0008-5472.CAN-08-1522 18794129

28. Li, Y., et al., Protein phosphatase 2A and DNA-dependent protein kinase are involved in mediating rapamycin-induced Akt phosphorylation. J Biol Chem, 2013.

29. Guertin D.A., et al., Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCalpha, but not S6K1. Dev Cell, 2006. 11(6): p. 859–71. 17141160

30. Meyer D.S., et al., Luminal expression of PIK3CA mutant H1047R in the mammary gland induces heterogeneous tumors. Cancer Res, 2011. 71(13): p. 4344–51. doi: 10.1158/0008-5472.CAN-10-3827 21482677

31. Boxer R.B., et al., Isoform-specific requirement for Akt1 in the developmental regulation of cellular metabolism during lactation. Cell Metab, 2006. 4(6): p. 475–90. 17141631

32. Chen C.C., et al., Akt is required for Stat5 activation and mammary differentiation. Breast Cancer Res, 2010. 12(5): p. R72. doi: 10.1186/bcr2640 20849614

33. Chen C.C., et al., Autocrine prolactin induced by the Pten-Akt pathway is required for lactation initiation and provides a direct link between the Akt and Stat5 pathways. Genes Dev, 2012. 26(19): p. 2154–68. doi: 10.1101/gad.197343.112 23028142

34. Debnath J., Walker S.J., and Brugge J.S., Akt activation disrupts mammary acinar architecture and enhances proliferation in an mTOR-dependent manner. J Cell Biol, 2003. 163(2): p. 315–26. 14568991

35. Zhang B., Zhang Y., and Shacter E., Rac1 inhibits apoptosis in human lymphoma cells by stimulating Bad phosphorylation on Ser-75. Mol Cell Biol, 2004. 24(14): p. 6205–14. 15226424

36. Zhu W. and Nelson C.M., PI3K regulates branch initiation and extension of cultured mammary epithelia via Akt and Rac1 respectively. Dev Biol, 2013. 379(2): p. 235–45. doi: 10.1016/j.ydbio.2013.04.029 23665174

37. Baselga J., Targeting the phosphoinositide-3 (PI3) kinase pathway in breast cancer. Oncologist, 2011. 16 Suppl 1: p. 12–9. doi: 10.1634/theoncologist.2011-S1-12 21278436

38. Pollack M.N., Insulin, insulin-like growth factors, insulin resistance, and neoplasia. Am J Clin Nutr, 2007. 86(3): p. s820–2. 18265475

39. Sachdev D. and Yee D., The IGF system and breast cancer. Endocr Relat Cancer, 2001. 8(3): p. 197–209. 11566611

40. Fenton T.R. and Gout I.T., Functions and regulation of the 70kDa ribosomal S6 kinases. Int J Biochem Cell Biol, 2011. 43(1): p. 47–59. doi: 10.1016/j.biocel.2010.09.018 20932932

41. Serra, V., et al., RSK3/4 mediate resistance to PI3K pathway inhibitors in breast cancer. J Clin Invest, 2013.

42. Miller T.W., Balko J.M., and Arteaga C.L., Phosphatidylinositol 3-kinase and antiestrogen resistance in breast cancer. J Clin Oncol, 2011. 29(33): p. 4452–61. doi: 10.1200/JCO.2010.34.4879 22010023

43. Miller T.W., et al., Mutations in the phosphatidylinositol 3-kinase pathway: role in tumor progression and therapeutic implications in breast cancer. Breast Cancer Res, 2011. 13(6): p. 224. doi: 10.1186/bcr3039 22114931

44. Li G., et al., Conditional loss of PTEN leads to precocious development and neoplasia in the mammary gland. Development, 2002. 129(17): p. 4159–70. 12163417

45. Wander S.A., Hennessy B.T., and Slingerland J.M., Next-generation mTOR inhibitors in clinical oncology: how pathway complexity informs therapeutic strategy. J Clin Invest, 2011. 121(4): p. 1231–41. doi: 10.1172/JCI44145 21490404

46. Janku F., et al., PIK3CA mutations in patients with advanced cancers treated with PI3K/AKT/mTOR axis inhibitors. Mol Cancer Ther, 2011. 10(3): p. 558–65. doi: 10.1158/1535-7163.MCT-10-0994 21216929

47. Janku F., et al., PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations. J Clin Oncol, 2012. 30(8): p. 777–82. doi: 10.1200/JCO.2011.36.1196 22271473

48. Janku F., et al., PIK3CA mutation H1047R is associated with response to PI3K/AKT/mTOR signaling pathway inhibitors in early-phase clinical trials. Cancer Res, 2013. 73(1): p. 276–84. doi: 10.1158/0008-5472.CAN-12-1726 23066039

49. Li H., et al., Targeting of mTORC2 prevents cell migration and promotes apoptosis in breast cancer. Breast Cancer Res Treat, 2012. 134(3): p. 1057–66. doi: 10.1007/s10549-012-2036-2 22476852

50. Hietakangas V. and Cohen S.M., TOR complex 2 is needed for cell cycle progression and anchorage-independent growth of MCF7 and PC3 tumor cells. BMC Cancer, 2008. 8: p. 282. doi: 10.1186/1471-2407-8-282 18831768

51. Wazir U., et al., Prognostic and therapeutic implications of mTORC1 and Rictor expression in human breast cancer. Oncol Rep, 2013. 29(5): p. 1969–74. doi: 10.3892/or.2013.2346 23503572

52. Serrano I., et al., Role of the integrin-linked kinase (ILK)/Rictor complex in TGFbeta-1-induced epithelial-mesenchymal transition (EMT). Oncogene, 2013. 32(1): p. 50–60. doi: 10.1038/onc.2012.30 22310280

53. Kim H.Y. and Nelson C.M., Extracellular matrix and cytoskeletal dynamics during branching morphogenesis. Organogenesis, 2012. 8(2): p. 56–64. doi: 10.4161/org.19813 22609561

54. Schnelzer A., et al., Rac1 in human breast cancer: overexpression, mutation analysis, and characterization of a new isoform, Rac1b. Oncogene, 2000. 19(26): p. 3013–20. 10871853

55. Fritz G., et al., Rho GTPases in human breast tumours: expression and mutation analyses and correlation with clinical parameters. Br J Cancer, 2002. 87(6): p. 635–44. 12237774

56. Katz E., et al., Targeting of Rac GTPases blocks the spread of intact human breast cancer. Oncotarget, 2012. 3(6): p. 608–19. 22689141

57. Brantley-Sieders D.M., et al., The receptor tyrosine kinase EphA2 promotes mammary adenocarcinoma tumorigenesis and metastatic progression in mice by amplifying ErbB2 signaling. J Clin Invest, 2008. 118(1): p. 64–78. 18079969

58. Qu S., et al., Gene targeting of ErbB3 using a Cre-mediated unidirectional DNA inversion strategy. Genesis, 2006. 44(10): p. 477–86. 16991114

59. Brantley-Sieders D.M., et al., EphA2 receptor tyrosine kinase regulates endothelial cell migration and vascular assembly through phosphoinositide 3-kinase-mediated Rac1 GTPase activation. J Cell Sci, 2004. 117(Pt 10): p. 2037–49. 15054110

60. Cheng N., et al., Transforming growth factor-beta signaling-deficient fibroblasts enhance hepatocyte growth factor signaling in mammary carcinoma cells to promote scattering and invasion. Mol Cancer Res, 2008. 6(10): p. 1521–33. doi: 10.1158/1541-7786.MCR-07-2203 18922968

61. Liang C.C., Park A.Y., and Guan J.L., In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc, 2007. 2(2): p. 329–33. 17406593

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 7
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#