The retinoblastoma tumor suppressor pathway modulates the invasiveness of ErbB2-positive breast cancer

A K Witkiewicz, D W Cox, D Rivadeneira, A E Ertel, P Fortina, G F Schwartz, E S Knudsen, A K Witkiewicz, D W Cox, D Rivadeneira, A E Ertel, P Fortina, G F Schwartz, E S Knudsen

Abstract

The processes that control the progression of ductal carcinoma in situ (DCIS) to invasive breast cancer remain poorly understood. Epidermal growth factor receptor 2 (ErbB2) overexpression is common in DCIS, as is disruption of the retinoblastoma tumor suppressor (RB) pathway. Here, we examined the cooperative impact of ErbB2 and RB deregulation on facets of disease progression. Our studies demonstrate that RB deficiency altered the expression of key molecules needed for proper cellular organization and epithelial cell-cell adhesion as part of a program related to the epithelial-to-mesenchymal transition (EMT). An increase in the invasive potential of ErbB2-overexpressing cells was observed upon RB depletion. Further, stable knockdown of RB resulted in invasive lesions in orthotopic xenograft assays, compared with DCIS-like lesions developing from RB-proficient cells. Conversely, the invasive phenotype observed in ErbB2-positive cancer models was inhibited through CDK4/6 inhibition in an RB-dependent manner. Finally, in a cohort of DCIS cases, we show that, although elevated levels of ErbB2 are associated with increased risk of a subsequent DCIS recurrence, it is not associated with progression to invasive disease. In contrast, RB loss in ErbB2-positive DCIS cases was associated with increased risk for invasive breast cancer. Taken together, these data demonstrate a key role for the RB pathway in invasion associated with breast tumor progression, and shed light on the key molecular events that promote the progression of DCIS to invasive disease.

Figures

Figure 1. RB-deficient or ErbB2 over expression…
Figure 1. RB-deficient or ErbB2 over expression promotes bypass of acini growth arrest
(A) Cells stably expressing miRB or miNS were analyzed by immunoblot for the indicated proteins (top panel). BrdU incorporation was analyzed by bivariate flow cytometry and data are the average of at least three independent experiments; bars, SD (bottom panel). (B) Ki67 staining (green) on 10 day acini, (40X) and Ki67 quantification from at least three independent experiments scoring a minimum of 20 acinus; bars, SD (***P < .0001). (C) Cells were infected with ErbB2 lentivirus and subsequently infected with miRB or miNS retroviruses. Cells were harvested and cell lysates were analyzed by immunoblot for the indicated proteins (top panel). BrdU incorporation was analyzed by bivariate flow cytometry and data are the average of at least three independent experiments; bars, SD (bottom panel). (D) Ki67 staining (green) on 10 day acini (40X) and Ki67 quantification from at least three independent experiments scoring a minimum of 20 acinus; bars, SD.
Figure 2. Combined effect of ErbB2- and…
Figure 2. Combined effect of ErbB2- and RB-pathway deregulation on acinar morphology
(A) Phase contrast images of acini (10X) were used to measure acini diameters and > 50 acini were scored for each genotype; bars, SD (***P < .0001) Phalloidin staining (red) was performed on acini grown for 10days (40x). (B) Phase contrast images of acini (20X) were used to visualize stellate acinar structures. Representative images are shown. (C) Gene expression data comparing MCF10A/ErbB2 miNS and MCF10A/ErbB2 miRB grown in 3D cultures. Changes in gene expression are displayed as a heat map (left panel). GSEA associated with the Taube-EMT signature (middle panel) and representative genes with corresponding fold change and P-values are highlighted (right panel).
Figure 2. Combined effect of ErbB2- and…
Figure 2. Combined effect of ErbB2- and RB-pathway deregulation on acinar morphology
(A) Phase contrast images of acini (10X) were used to measure acini diameters and > 50 acini were scored for each genotype; bars, SD (***P < .0001) Phalloidin staining (red) was performed on acini grown for 10days (40x). (B) Phase contrast images of acini (20X) were used to visualize stellate acinar structures. Representative images are shown. (C) Gene expression data comparing MCF10A/ErbB2 miNS and MCF10A/ErbB2 miRB grown in 3D cultures. Changes in gene expression are displayed as a heat map (left panel). GSEA associated with the Taube-EMT signature (middle panel) and representative genes with corresponding fold change and P-values are highlighted (right panel).
Figure 3. RB loss compromises the integrity…
Figure 3. RB loss compromises the integrity of cell-cell adhesion complexes
(A) Immunoblot analysis of MCF10A ErbB2 cells either transduced with miNS or miRB are shown for the indicated proteins. (B) Representative images of cells in 2D cultured stained for the indicated cytokeratins. (C) Representative images of CK18 stained in 3D cultures. (D) Representative images of cells in 2D stained for junctional plakoglobin (JUP). (E) Representative images of cell in 2D cultured stained for E-cadherin (green) and Phalloidin (red) (top panel) and acini stained for E-cadherin (red) are shown. Leading edges of RB-deficient cells displaying decreased E-cadherin expression are highlighted (arrows, bottom panel). (F) Representative images for Laminin V in 3D cultures.
Figure 4. RB loss promotes cell migration…
Figure 4. RB loss promotes cell migration and invasion in ErbB2 over expressing cells
(A) Cell migration and (B) cell invasion of MCF10A cells harboring ErbB2 over expression and/or RB-deficiency by Boyden Chamber assays; bars, SD. (***P < .0001) (C/D) SKBR3 and BT474 cells transduced with miRB or miNS retroviruses. Cells were harvested and cell lysates were analyzed by immunoblot for the indicated proteins. BrdU incorporation was analyzed by bivariate flow cytometry and data are average of at least three independent experiments; bars, SD. (E/F) Analysis of SKBR3 miNS/miRB and BT474 miNS/miRB cell migration by Boyden Chamber assays; bars, SD. (**P<0.01) (F) Analysis of SKBR3 miNS/miRB and BT474 miNS/miRB cell invasion by Boyden Chamber assays; bars, SD. (**P<0.01).
Figure 5. Activation of the RB pathway…
Figure 5. Activation of the RB pathway prevents abnormal multi-acini growth and cell invasion
(A) RB-proficient MCF10A/ErbB2 cells were treated with 0.5μM PD-0332991 for 24h. Cells were harvested and cell lysates were analyzed by immunoblot for the indicated proteins (left panel). BrdU incorporation was analyzed by bivariate flow cytometry and data are the average of at least three independent experiments; bars, SD (***P < .0001) (middle panel). Ki67 and phalloidin co-staining of MCF10A/ErbB2 acini treated with 0.5μM PD-0332991 for 72h (20X) (right panel). (B) RB-proficient MCF10A/ErbB2 cells grown in 3D culture and treated with 0.5μM PD0332991 for 72h, (20X) (left panel). Analysis of cell migration by Boyden Chamber assays supplemented with 0.5μM PD0332991 was performed; bars, SD. (**P=.003) (right panel). (C) RB-proficient BT474 cells were treated with PD-0332991 and analyzed by immunoblotting, BrdU incorporation and proliferation in 3D culture. BrdU incorporation data are the average of at least three independent experiments; bars, SD (***P < .0001). (D) RB-proficient BT474 cells were treated for growth in 3D culture and cell migration. Migration data are the average of at least three independent experiments; bars, SD (**P < 0.0037).
Figure 6. RB plays a role in…
Figure 6. RB plays a role in preventing invasive tumor growth in vivo
(A) Dual staining for ErbB2 (red) and SMA (green) in SKBR3 miNS and SKBR3 miRB injected mammary glands. (B) Representative H&E, Smooth muscle actin (SMA) and Ki67 staining in SKBR3 miNS and SKBR3 miRB lesions are shown (left panels). Table depicting percentage of DCIS and invasive lesions from SKBR3 miNS and SKBR3 miRB orthotopic injections (n=15) (right panel)
Figure 7. RB deficiency is associated with…
Figure 7. RB deficiency is associated with increased disease progression in DCIS
(A) ErbB2 high (3+) status was compared against all other ErbB2 staining (0, 1+, 2+) for any recurrent disease (ipsilateral breast event) or invasive progression using Kaplan-Meier analysis. ErbB2 high was associated with risk of a subsequent ipsilateral breast event, but not progression to invasive disease. (B) Representative images of RB staining in DCIS lesions cases with high ErbB2. (C) ErbB2 high (3+) cases were stratified by RB status (positive vs. negative). Cases with RB loss in this subtype were associated with both risk of a subsequent ipsilateral breast event and progression to invasive disease.

References

    1. Ernster VL, Ballard-Barbash R, Barlow WE, Zheng Y, Weaver DL, Cutter G, et al. Detection of ductal carcinoma in situ in women undergoing screening mammography. J Natl Cancer Inst. 2002 Oct 16;94(20):1546–54.
    1. Virnig BA, Tuttle TM, Shamliyan T, Kane RL. Ductal carcinoma in situ of the breast: a systematic review of incidence, treatment, and outcomes. J Natl Cancer Inst. 2010 Feb 3;102(3):170–8. Epub 2010/01/15. eng.
    1. Betsill WL, Jr, Rosen PP, Lieberman PH, Robbins GF. Intraductal carcinoma. Long-term follow-up after treatment by biopsy alone. Jama. 1978 May 5;239(18):1863–7.
    1. Carlson RW, Allred DC, Anderson BO, Burstein HJ, Carter WB, Edge SB, et al. Breast cancer. Clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2009 Feb;7(2):122–92. eng.
    1. Schnitt SJ. Local outcomes in ductal carcinoma in situ based on patient and tumor characteristics. J Natl Cancer Inst Monogr. 2010;(41):158–61. Epub 2010/10/20. eng.
    1. Allegra CJ, Aberle DR, Ganschow P, Hahn SM, Lee CN, Millon-Underwood S, et al. National Institutes of Health State-of-the-Science Conference statement: Diagnosis and Management of Ductal Carcinoma In Situ September 22-24, 2009. J Natl Cancer Inst. Feb 3;102(3):161–9. eng.
    1. Jones JL. Overdiagnosis and overtreatment of breast cancer: progression of ductal carcinoma in situ: the pathological perspective. Breast Cancer Res. 2006;8(2):204.
    1. Romero L, Klein L, Ye W, Holmes D, Soni R, Silberman H, et al. Outcome after invasive recurrence in patients with ductal carcinoma in situ of the breast. Am J Surg. 2004 Oct;188(4):371–6.
    1. Kerlikowske K, Molinaro A, Cha I, Ljung BM, Ernster VL, Stewart K, et al. Characteristics associated with recurrence among women with ductal carcinoma in situ treated by lumpectomy. J Natl Cancer Inst. 2003 Nov 19;95(22):1692–702.
    1. Knudsen ES, Ertel A, Davicioni E, Kline J, Schwartz GF, Witkiewicz AK. Progression of ductal carcinoma in situ to invasive breast cancer is associated with gene expression programs of EMT and myoepithelia. Breast Cancer Res Treat. 2011 Jun;133(3):1009–24. Epub 2011/12/03. eng.
    1. Lee S, Stewart S, Nagtegaal I, Luo J, Wu Y, Colditz G, et al. Differentially expressed genes regulating the progression of ductal carcinoma in situ to invasive breast cancer. Cancer research. 2012 Sep 1;72(17):4574–86.
    1. Franke WW. Discovering the molecular components of intercellular junctions--a historical view. Cold Spring Harb Perspect Biol. 2009 Sep;1(3):a003061.
    1. Zhurinsky J, Shtutman M, Ben-Ze'ev A. Plakoglobin and beta-catenin: protein interactions, regulation and biological roles. J Cell Sci. 2000 Sep;113(Pt 18):3127–39.
    1. Wheelock MJ, Johnson KR. Cadherins as modulators of cellular phenotype. Annu Rev Cell Dev Biol. 2003;19:207–35.
    1. Rodriguez-Boulan E, Nelson WJ. Morphogenesis of the polarized epithelial cell phenotype. Science. 1989 Aug 18;245(4919):718–25.
    1. O'Brien LE, Zegers MM, Mostov KE. Opinion: Building epithelial architecture: insights from three-dimensional culture models. Nat Rev Mol Cell Biol. 2002 Jul;3(7):531–7.
    1. Kim YS, Won YS, Park KS, Song BJ, Kim JS, Oh SJ, et al. Prognostic significance of HER2 gene amplification according to stage of breast cancer. J Korean Med Sci. 2008 Jun;23(3):414–20.
    1. Ross JS, Slodkowska EA, Symmans WF, Pusztai L, Ravdin PM, Hortobagyi GN. The HER-2 receptor and breast cancer: ten years of targeted anti-HER-2 therapy and personalized medicine. Oncologist. 2009 Apr;14(4):320–68.
    1. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987 Jan 9;235(4785):177–82. eng.
    1. Allred DC, Clark GM, Molina R, Tandon AK, Schnitt SJ, Gilchrist KW, et al. Overexpression of HER-2/neu and its relationship with other prognostic factors change during the progression of in situ to invasive breast cancer. Hum Pathol. 1992 Sep;23(9):974–9. eng.
    1. Hoque A, Sneige N, Sahin AA, Menter DG, Bacus JW, Hortobagyi GN, et al. Her-2/neu gene amplification in ductal carcinoma in situ of the breast. Cancer Epidemiol Biomarkers Prev. 2002 Jun;11(6):587–90. eng.
    1. Muthuswamy SK, Li D, Lelievre S, Bissell MJ, Brugge JS. ErbB2, but not ErbB1, reinitiates proliferation and induces luminal repopulation in epithelial acini. Nat Cell Biol. 2001 Sep;3(9):785–92. eng.
    1. Wu M, Jung L, Cooper AB, Fleet C, Chen L, Breault L, et al. Dissecting genetic requirements of human breast tumorigenesis in a tissue transgenic model of human breast cancer in mice. Proc Natl Acad Sci U S A. 2009 Apr 28;106(17):7022–7. eng.
    1. Kerlikowske K, Molinaro AM, Gauthier ML, Berman HK, Waldman F, Bennington J, et al. Biomarker expression and risk of subsequent tumors after initial ductal carcinoma in situ diagnosis. J Natl Cancer Inst. 2010 May 5;102(9):627–37. eng.
    1. Gauthier ML, Berman HK, Miller C, Kozakeiwicz K, Chew K, Moore D, et al. Abrogated response to cellular stress identifies DCIS associated with subsequent tumor events and defines basal-like breast tumors. Cancer Cell. 2007 Nov;12(5):479–91. eng.
    1. Witkiewicz AK, Rivadeneira DB, Ertel A, Kline J, Hyslop T, Schwartz GF, et al. Association of RB/p16-Pathway Perturbations with DCIS Recurrence Dependence on Tumor versus Tissue Microenvironment. Am J Pathol. 2010 Sep;179(3):1171–8. eng.
    1. Knudsen ES, Pajak TF, Qeenan M, McClendon AK, Armon BD, Schwartz GF, et al. Retinoblastoma and phosphate and tensin homolog tumor suppressors: impact on ductal carcinoma in situ progression. Journal of the National Cancer Institute. 2012 Dec 5;104(23):1825–36.
    1. Knudsen ES, Knudsen KE. Tailoring to RB: tumour suppressor status and therapeutic response. Nat Rev Cancer. 2008 Sep;8(9):714–24. eng.
    1. Cobrinik D. Pocket proteins and cell cycle control. Oncogene. 2005 Apr 18;24(17):2796–809.
    1. Jiang Z, Deng T, Jones R, Li H, Herschkowitz JI, Liu JC, et al. Rb deletion in mouse mammary progenitors induces luminal-B or basal-like/EMT tumor subtypes depending on p53 status. J Clin Invest. 2010 Sep 1;120(9):3296–309. eng.
    1. Herschkowitz JI, He X, Fan C, Perou CM. The functional loss of the retinoblastoma tumour suppressor is a common event in basal-like and luminal B breast carcinomas. Breast Cancer Res. 2008 Sep 9;10(5):R75. Epub 2008/09/11. Eng.
    1. Debnath J, Muthuswamy SK, Brugge JS. Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures. Methods. 2003 Jul;30(3):256–68. Epub 2003/06/12. eng.
    1. Hebner C, Weaver VM, Debnath J. Modeling morphogenesis and oncogenesis in three-dimensional breast epithelial cultures. Annu Rev Pathol. 2008;3:313–39.
    1. Kenny PA, Lee GY, Myers CA, Neve RM, Semeiks JR, Spellman PT, et al. The morphologies of breast cancer cell lines in three-dimensional assays correlate with their profiles of gene expression. Mol Oncol. 2007 Jun;1(1):84–96. Epub 2008/06/03. eng.
    1. Taube JH, Herschkowitz JI, Komurov K, Zhou AY, Gupta S, Yang J, et al. Core epithelial-to-mesenchymal transition interactome gene-expression signature is associated with claudin-low and metaplastic breast cancer subtypes. Proc Natl Acad Sci U S A. 2010 Aug 31;107(35):15449–54. eng.
    1. Petersen OW, Ronnov-Jessen L, Howlett AR, Bissell MJ. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proc Natl Acad Sci U S A. 1992 Oct 1;89(19):9064–8.
    1. Toogood PL, Harvey PJ, Repine JT, Sheehan DJ, VanderWel SN, Zhou H, et al. Discovery of a potent and selective inhibitor of cyclin-dependent kinase 4/6. J Med Chem. 2005 Apr 7;48(7):2388–406.
    1. Fry DW, Harvey PJ, Keller PR, Elliott WL, Meade M, Trachet E, et al. Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts. Mol Cancer Ther. 2004 Nov;3(11):1427–38.
    1. Dean JL, Thangavel C, McClendon AK, Reed CA, Knudsen ES. Therapeutic CDK4/6 inhibition in breast cancer: key mechanisms of response and failure. Oncogene. 2010 Jul 15;29(28):4018–32. eng.
    1. Zajchowski DA, Bartholdi MF, Gong Y, Webster L, Liu HL, Munishkin A, et al. Identification of gene expression profiles that predict the aggressive behavior of breast cancer cells. Cancer Res. 2001 Jul 1;61(13):5168–78.
    1. Rakovitch E, Nofech-Mozes S, Hanna W, Narod S, Thiruchelvam D, Saskin R, et al. HER2/neu and Ki-67 expression predict non-invasive recurrence following breast-conserving therapy for ductal carcinoma in situ. British journal of cancer. 2012 Mar 13;106(6):1160–5.
    1. Correa C, McGale P, Taylor C, Wang Y, Clarke M, Davies C, et al. Overview of the randomized trials of radiotherapy in ductal carcinoma in situ of the breast. J Natl Cancer Inst Monogr. 2010;(41):162–77. Epub 2010/10/20. eng.
    1. Erbas B, Provenzano E, Armes J, Gertig D. The natural history of ductal carcinoma in situ of the breast: a review. Breast Cancer Res Treat. 2006 May;97(2):135–44. Epub 2005/12/02. eng.
    1. Leonard GD, Swain SM. Ductal carcinoma in situ, complexities and challenges. Journal of the National Cancer Institute. 2004 Jun 16;96(12):906–20. eng.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2012 Mar 4;144(5):646–74.
    1. Cavallaro U, Christofori G. Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer. 2004 Feb;4(2):118–32.
    1. Knudsen ES, Ertel A, Davicioni E, Kline J, Schwartz GF, Witkiewicz AK. Progression of ductal carcinoma in situ to invasive breast cancer is associated with gene expression programs of EMT and myoepithelia. Breast Cancer Res Treat. 2011 Dec 2; Epub 2011/12/03. Eng.
    1. Sutherland RL, Musgrove EA. Cyclins and breast cancer. J Mammary Gland Biol Neoplasia. 2004 Jan;9(1):95–104.
    1. Rivadeneira DB, Mayhew CN, Thangavel C, Sotillo E, Reed CA, Grana X, et al. Proliferative suppression by CDK4/6 inhibition: complex function of the retinoblastoma pathway in liver tissue and hepatoma cells. Gastroenterology. 2010 May;138(5):1920–30.
    1. Thangavel C, Dean JL, Ertel A, Knudsen KE, Aldaz CM, Witkiewicz AK, et al. Therapeutically activating RB: reestablishing cell cycle control in endocrine therapy-resistant breast cancer. Endocr Relat Cancer. 2011;18(3):333–45. eng.
    1. Knudsen ES, Buckmaster C, Chen TT, Feramisco JR, Wang JY. Inhibition of DNA synthesis by RB: effects on G1/S transition and S-phase progression. Genes Dev. 1998 Aug 1;12(15):2278–92.
    1. Sotgia F, Williams TM, Schubert W, Medina F, Minetti C, Pestell RG, et al. Caveolin-1 deficiency (-/-) conveys premalignant alterations in mammary epithelia, with abnormal lumen formation, growth factor independence, and cell invasiveness. Am J Pathol. 2006 Jan;168(1):292–309. Epub 2006/01/10. eng.
    1. Storey JD. A direct approach to false discovery rates. Journal of the Royal Statistical Society: Series B (Statistical Methodology) 2002;64(3):479–98.
    1. Hackstadt AJ, Hess AM. Filtering for increased power for microarray data analysis. BMC Bioinformatics. 2009;10:11. Epub 2009/01/10. eng.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005 Oct 25;102(43):15545–50.
    1. McClendon AK, Dean JL, Ertel A, Fu Z, Rivadeneira DB, Reed CA, et al. RB and p53 cooperate to prevent liver tumorigenesis in response to tissue damage. Gastroenterology. 2011 Oct;141(4):1439–50.

Source: PubMed

3
購読する