Skip to main content
Erschienen in: Clinical & Experimental Metastasis 2/2017

17.02.2017 | Research Paper

Astrocyte-induced Reelin expression drives proliferation of Her2+ breast cancer metastases

verfasst von: Rahul Jandial, Cecilia Choy, Danielle M Levy, Mike Y Chen, Khairul I Ansari

Erschienen in: Clinical & Experimental Metastasis | Ausgabe 2/2017

Einloggen, um Zugang zu erhalten

Abstract

Breast cancer metastasis to the brain develops after a clinical latency of years to even decades, suggesting that colonization of the brain is the most challenging step of the metastatic cascade. However, the underlying mechanisms used by breast cancer cells to successfully colonize the brain’s microenvironment remain elusive. Reelin is an archetypal extracellular glycoprotein that regulates migration, proliferation, and lamination of neurons. It is epigenetically silenced in various cancers, and its expression in multiple myelomas is linked to poor patient survival. We found that Reelin expression was low in primary breast cancer tissue. However, its expression was significantly higher in Her2+ breast cancers metastasizing to the brain. In particular, Reelin was highly expressed in the tumor periphery adjacent to surrounding astrocytes. This augmented Reelin expression was seen in Her2+ metastases, but not in triple negative (TN) primary tumors or in TN breast to brain metastasis cells co-cultured with astrocytes. Furthermore, the elevated expression was sustained in Her2+ cells grown in the presence of the DNA methyltransferase inhibitor 5-azacytidine, indicating epigenetic regulation of Reelin expression. The relative growth and rate of spheroids formation derived from Her2+ primary and BBM cells co-cultured with astrocytes were higher than those of TN primary and BBM cells, and knockdown of both Reelin and Her2 suppressed the astrocyte-induced growth and spheroid forming ability of Her2+ cells. Collectively, our results indicate that within the neural niche, astrocytes epigenetically regulate Reelin expression and its interaction with Her2 leading to increased proliferation and survival fitness.
Literatur
1.
Zurück zum Zitat Kondziolka D, Kalkanis SN, Mehta MP, Ahluwalia M, Loeffler JS (2014) It is time to reevaluate the management of patients with brain metastases. Neurosurgery 75:1–9CrossRefPubMed Kondziolka D, Kalkanis SN, Mehta MP, Ahluwalia M, Loeffler JS (2014) It is time to reevaluate the management of patients with brain metastases. Neurosurgery 75:1–9CrossRefPubMed
2.
Zurück zum Zitat Honda Y, Goto R, Idera N, Horiguchi K, Kitagawa D, et al. (2014) Prognostic significance of subtypes and Gpa (Graded Prognostic Assessment) in brain metastases from breast cancer. Ann Oncol Honda Y, Goto R, Idera N, Horiguchi K, Kitagawa D, et al. (2014) Prognostic significance of subtypes and Gpa (Graded Prognostic Assessment) in brain metastases from breast cancer. Ann Oncol
3.
4.
Zurück zum Zitat Percy DB, Ribot EJ, Chen Y, McFadden C, Simedrea C et al (2011) In vivo characterization of changing blood-tumor barrier permeability in a mouse model of breast cancer metastasis: a complementary magnetic resonance imaging approach. Invest Radiol 46:718–725PubMed Percy DB, Ribot EJ, Chen Y, McFadden C, Simedrea C et al (2011) In vivo characterization of changing blood-tumor barrier permeability in a mouse model of breast cancer metastasis: a complementary magnetic resonance imaging approach. Invest Radiol 46:718–725PubMed
5.
Zurück zum Zitat Piccirilli M, Sassun TE, Brogna C, Giangaspero F, Salvati M (2007) Late brain metastases from breast cancer: clinical remarks on 11 patients and review of the literature. Tumori 93:150–154PubMed Piccirilli M, Sassun TE, Brogna C, Giangaspero F, Salvati M (2007) Late brain metastases from breast cancer: clinical remarks on 11 patients and review of the literature. Tumori 93:150–154PubMed
7.
Zurück zum Zitat Jandial R, Anderson A, Choy C, Levy ML (2012) Bidirectional microevnironmental cues between neoplastic and stromal cells drive metastasis formation and efficiency. Neurosurgery 70:N12–N13.CrossRefPubMed Jandial R, Anderson A, Choy C, Levy ML (2012) Bidirectional microevnironmental cues between neoplastic and stromal cells drive metastasis formation and efficiency. Neurosurgery 70:N12–N13.CrossRefPubMed
9.
Zurück zum Zitat Langley RR, Fidler IJ (2011) The seed and soil hypothesis revisited-The role of tumor-stroma interactions in metastasis to different organs. Int J Cancer 128:2527–2535CrossRefPubMedPubMedCentral Langley RR, Fidler IJ (2011) The seed and soil hypothesis revisited-The role of tumor-stroma interactions in metastasis to different organs. Int J Cancer 128:2527–2535CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Curran T, D’Arcangelo G (1998) Role of Reelin in the control of brain development. Brain Res Rev 26:285–294CrossRefPubMed Curran T, D’Arcangelo G (1998) Role of Reelin in the control of brain development. Brain Res Rev 26:285–294CrossRefPubMed
11.
Zurück zum Zitat Rice DS, Curran T (2001) Role of the Reelin signaling pathway in central nervous system development. Annu Rev Neurosci 24:1005–1039CrossRefPubMed Rice DS, Curran T (2001) Role of the Reelin signaling pathway in central nervous system development. Annu Rev Neurosci 24:1005–1039CrossRefPubMed
12.
Zurück zum Zitat Zhou Q, Obana EA, Radomski KL, Sukumar G, Wynder C et al (2016) Inhibition of the histone demethylase Kdm5b promotes neurogenesis and derepresses Reln (reelin) in neural stem cells from the adult subventricular zone of mice. Mol Biol Cell 27:627–639CrossRefPubMedPubMedCentral Zhou Q, Obana EA, Radomski KL, Sukumar G, Wynder C et al (2016) Inhibition of the histone demethylase Kdm5b promotes neurogenesis and derepresses Reln (reelin) in neural stem cells from the adult subventricular zone of mice. Mol Biol Cell 27:627–639CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Mitchell CP, Chen Y, Kundakovic M, Costa E, Grayson DR (2005) Histone deacetylase inhibitors decrease reelin promoter methylation in vitro. J Neurochem 93:483–492CrossRefPubMed Mitchell CP, Chen Y, Kundakovic M, Costa E, Grayson DR (2005) Histone deacetylase inhibitors decrease reelin promoter methylation in vitro. J Neurochem 93:483–492CrossRefPubMed
14.
Zurück zum Zitat Stein T, Cosimo E, Yu X, Smith PR, Simon R et al (2010) Loss of reelin expression in breast cancer is epigenetically controlled and associated with poor prognosis. Am J Pathol 177:2323–2333CrossRefPubMedPubMedCentral Stein T, Cosimo E, Yu X, Smith PR, Simon R et al (2010) Loss of reelin expression in breast cancer is epigenetically controlled and associated with poor prognosis. Am J Pathol 177:2323–2333CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Neman J, Termini J, Wilczynski S, Vaidehi N, Choy C et al (2014) Human breast cancer metastases to the brain display GABAergic properties in the neural niche. Proc Natl Acad Sci USA 111:984–989CrossRefPubMedPubMedCentral Neman J, Termini J, Wilczynski S, Vaidehi N, Choy C et al (2014) Human breast cancer metastases to the brain display GABAergic properties in the neural niche. Proc Natl Acad Sci USA 111:984–989CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Zhang Y, Sloan SA, Clarke LE, Caneda C, Plaza CA et al (2016) Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89:37–53CrossRefPubMed Zhang Y, Sloan SA, Clarke LE, Caneda C, Plaza CA et al (2016) Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89:37–53CrossRefPubMed
17.
Zurück zum Zitat Raposo G, Nijman HW, Stoorvogel W, Leijendekker R, Harding CV et al (1996) B lymphocytes secrete antigen-presenting vesicles. J Exp Med 183:1161–1172CrossRefPubMed Raposo G, Nijman HW, Stoorvogel W, Leijendekker R, Harding CV et al (1996) B lymphocytes secrete antigen-presenting vesicles. J Exp Med 183:1161–1172CrossRefPubMed
18.
Zurück zum Zitat Bronisz A, Wang Y, Nowicki MO, Peruzzi P, Ansari KI et al (2014) Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1. Cancer Res 74:738–750CrossRefPubMed Bronisz A, Wang Y, Nowicki MO, Peruzzi P, Ansari KI et al (2014) Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1. Cancer Res 74:738–750CrossRefPubMed
19.
Zurück zum Zitat Neman J, Choy C, Kowolik CM, Anderson A, Duenas VJ et al (2013) Co-evolution of breast-to-brain metastasis and neural progenitor cells. Clin Exp Metastasis 30:753–768CrossRefPubMed Neman J, Choy C, Kowolik CM, Anderson A, Duenas VJ et al (2013) Co-evolution of breast-to-brain metastasis and neural progenitor cells. Clin Exp Metastasis 30:753–768CrossRefPubMed
20.
Zurück zum Zitat Massalini S, Pellegatta S, Pisati F, Finocchiaro G, Farace MG et al (2009) Reelin affects chain-migration and differentiation of neural precursor cells. Mol Cell Neurosci 42:341–349CrossRefPubMed Massalini S, Pellegatta S, Pisati F, Finocchiaro G, Farace MG et al (2009) Reelin affects chain-migration and differentiation of neural precursor cells. Mol Cell Neurosci 42:341–349CrossRefPubMed
21.
Zurück zum Zitat Becker J, Frohlich J, Perske C, Pavlakovic H, Wilting J (2012) Reelin signalling in neuroblastoma: migratory switch in metastatic stages. Int J Oncol 41:681–689PubMed Becker J, Frohlich J, Perske C, Pavlakovic H, Wilting J (2012) Reelin signalling in neuroblastoma: migratory switch in metastatic stages. Int J Oncol 41:681–689PubMed
22.
Zurück zum Zitat Jossin Y, Cooper JA (2011) Reelin, Rap1 and N-cadherin orient the migration of multipolar neurons in the developing neocortex. Nat Neurosci 14:697–703CrossRefPubMedPubMedCentral Jossin Y, Cooper JA (2011) Reelin, Rap1 and N-cadherin orient the migration of multipolar neurons in the developing neocortex. Nat Neurosci 14:697–703CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat D’Arcangelo G (2014) Reelin in the years: controlling neuronal migration and maturation in the mammalian brain. Adv Neurosci 2014:19 D’Arcangelo G (2014) Reelin in the years: controlling neuronal migration and maturation in the mammalian brain. Adv Neurosci 2014:19
24.
Zurück zum Zitat Biamonte F, Scicchitano BB, Lama G (2015) Evaluation of the Reelin signaling in cancer stem cells isolated from tumor and peritumor tissue of glioblastoma. Ital J Anat Embriol 120(1):89. Biamonte F, Scicchitano BB, Lama G (2015) Evaluation of the Reelin signaling in cancer stem cells isolated from tumor and peritumor tissue of glioblastoma. Ital J Anat Embriol 120(1):89.
25.
Zurück zum Zitat Evangelisti C, Florian MC, Massimi I, Dominici C, Giannini G et al (2009) MiR-128 up-regulation inhibits Reelin and DCX expression and reduces neuroblastoma cell motility and invasiveness. FASEB J 23:4276–4287CrossRefPubMed Evangelisti C, Florian MC, Massimi I, Dominici C, Giannini G et al (2009) MiR-128 up-regulation inhibits Reelin and DCX expression and reduces neuroblastoma cell motility and invasiveness. FASEB J 23:4276–4287CrossRefPubMed
26.
Zurück zum Zitat Hu HL, Deng F, Liu Y, Chen MF, Zhang XL et al (2012) Characterization and retinal neuron differentiation of WERI-Rb1 cancer stem cells. Mol Vision 18:2388–2397 Hu HL, Deng F, Liu Y, Chen MF, Zhang XL et al (2012) Characterization and retinal neuron differentiation of WERI-Rb1 cancer stem cells. Mol Vision 18:2388–2397
27.
Zurück zum Zitat Seigel GM, Hackam AS, Ganguly A, Mandell LM, Gonzalez-Fernandez F (2007) Human embryonic and neuronal stem cell markers in retinoblastoma. Mol Vis 13:823–832PubMedPubMedCentral Seigel GM, Hackam AS, Ganguly A, Mandell LM, Gonzalez-Fernandez F (2007) Human embryonic and neuronal stem cell markers in retinoblastoma. Mol Vis 13:823–832PubMedPubMedCentral
28.
Zurück zum Zitat Lin L, Yan F, Zhao D, Lv M, Liang X et al (2016) Reelin promotes the adhesion and drug resistance of multiple myeloma cells via integrin beta1 signaling and STAT3. Oncotarget 7:9844–9858PubMedPubMedCentral Lin L, Yan F, Zhao D, Lv M, Liang X et al (2016) Reelin promotes the adhesion and drug resistance of multiple myeloma cells via integrin beta1 signaling and STAT3. Oncotarget 7:9844–9858PubMedPubMedCentral
29.
Zurück zum Zitat Sui L, Wang Y, Ju LH, Chen M (2012) Epigenetic regulation of reelin and brain-derived neurotrophic factor genes in long-term potentiation in rat medial prefrontal cortex. Neurobiol Learn Mem 97:425–440CrossRefPubMed Sui L, Wang Y, Ju LH, Chen M (2012) Epigenetic regulation of reelin and brain-derived neurotrophic factor genes in long-term potentiation in rat medial prefrontal cortex. Neurobiol Learn Mem 97:425–440CrossRefPubMed
30.
Zurück zum Zitat Dohi O, Takada H, Wakabayashi N, Yasui K, Sakakura C et al (2010) Epigenetic silencing of RELN in gastric cancer. Int J Oncol 36:85–92PubMed Dohi O, Takada H, Wakabayashi N, Yasui K, Sakakura C et al (2010) Epigenetic silencing of RELN in gastric cancer. Int J Oncol 36:85–92PubMed
31.
Zurück zum Zitat Grayson DR, Chen Y, Costa E, Dong E, Guidotti A et al (2006) The human reelin gene: transcription factors (+), repressors (-) and the methylation switch (+/-) in schizophrenia. Pharmacol Ther 111:272–286CrossRefPubMed Grayson DR, Chen Y, Costa E, Dong E, Guidotti A et al (2006) The human reelin gene: transcription factors (+), repressors (-) and the methylation switch (+/-) in schizophrenia. Pharmacol Ther 111:272–286CrossRefPubMed
32.
Zurück zum Zitat Biamonte F, Scicchitano BM, Lama G (2015) Evaluation of the Reelin signaling in cancer stem cells isolated from tumor and peritumor tissue of glioblastoma. Ital J Antomy Embryol 120:89 Biamonte F, Scicchitano BM, Lama G (2015) Evaluation of the Reelin signaling in cancer stem cells isolated from tumor and peritumor tissue of glioblastoma. Ital J Antomy Embryol 120:89
34.
35.
Zurück zum Zitat Rajendran L, Bali J, Barr MM, Court FA, Kramer-Albers EM et al (2014) Emerging roles of extracellular vesicles in the nervous system. J Neurosci 34:15482–15489CrossRefPubMedPubMedCentral Rajendran L, Bali J, Barr MM, Court FA, Kramer-Albers EM et al (2014) Emerging roles of extracellular vesicles in the nervous system. J Neurosci 34:15482–15489CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Zhang L, Zhang S, Yao J, Lowery FJ, Zhang Q et al (2015) Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature 527:100–104CrossRefPubMedPubMedCentral Zhang L, Zhang S, Yao J, Lowery FJ, Zhang Q et al (2015) Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature 527:100–104CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Liu HY, Huang CM, Hung YF, Hsueh YP (2015) The microRNAs Let7c and miR21 are recognized by neuronal Toll-like receptor 7 to restrict dendritic growth of neurons. Exp Neurol 269:202–212CrossRefPubMed Liu HY, Huang CM, Hung YF, Hsueh YP (2015) The microRNAs Let7c and miR21 are recognized by neuronal Toll-like receptor 7 to restrict dendritic growth of neurons. Exp Neurol 269:202–212CrossRefPubMed
38.
Zurück zum Zitat Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M et al (2015) Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol 17:183–194CrossRefPubMedPubMedCentral Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M et al (2015) Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol 17:183–194CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Lee SA, Lee H, Pinney JR, Khialeeva E, Bergsneider M et al (2011) Development of microfabricated magnetic actuators for removing cellular occlusion. J Micromech Microeng 21:54006CrossRefPubMed Lee SA, Lee H, Pinney JR, Khialeeva E, Bergsneider M et al (2011) Development of microfabricated magnetic actuators for removing cellular occlusion. J Micromech Microeng 21:54006CrossRefPubMed
40.
41.
Zurück zum Zitat Yuan Y, Chen H, Ma G, Cao X, Liu Z (2012) Reelin is involved in transforming growth factor-beta1-induced cell migration in esophageal carcinoma cells. PLoS ONE 7:e31802CrossRefPubMedPubMedCentral Yuan Y, Chen H, Ma G, Cao X, Liu Z (2012) Reelin is involved in transforming growth factor-beta1-induced cell migration in esophageal carcinoma cells. PLoS ONE 7:e31802CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Perrone G, Vincenzi B, Zagami M, Santini D, Panteri R et al (2007) Reelin expression in human prostate cancer: a marker of tumor aggressiveness based on correlation with grade. Mod Pathol 20:344–351CrossRefPubMed Perrone G, Vincenzi B, Zagami M, Santini D, Panteri R et al (2007) Reelin expression in human prostate cancer: a marker of tumor aggressiveness based on correlation with grade. Mod Pathol 20:344–351CrossRefPubMed
43.
Zurück zum Zitat Khialeeva E, Chou JW, Carpenter EM (2015) Reelin signaling in tumor microenvironment is required for metastasis of mammary carcinoma cells. Cancer Res. doi:10.1158/1538-7445 Khialeeva E, Chou JW, Carpenter EM (2015) Reelin signaling in tumor microenvironment is required for metastasis of mammary carcinoma cells. Cancer Res. doi:10.​1158/​1538-7445
44.
Zurück zum Zitat Khialeeva E, Chou JW, Carpenter EM (2015) Exploring the relationship between reelin signaling and breast cancer metastasis. Cancer Res. doi:10.1158/1538-7445 Khialeeva E, Chou JW, Carpenter EM (2015) Exploring the relationship between reelin signaling and breast cancer metastasis. Cancer Res. doi:10.​1158/​1538-7445
Metadaten
Titel
Astrocyte-induced Reelin expression drives proliferation of Her2+ breast cancer metastases
verfasst von
Rahul Jandial
Cecilia Choy
Danielle M Levy
Mike Y Chen
Khairul I Ansari
Publikationsdatum
17.02.2017
Verlag
Springer Netherlands
Erschienen in
Clinical & Experimental Metastasis / Ausgabe 2/2017
Print ISSN: 0262-0898
Elektronische ISSN: 1573-7276
DOI
https://doi.org/10.1007/s10585-017-9839-9

Weitere Artikel der Ausgabe 2/2017

Clinical & Experimental Metastasis 2/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.