Skip to main content
Top
Published in: Cancer Cell International 1/2016

Open Access 01-12-2016 | Primary Research

The availability of the embryonic TGF-β protein Nodal is dynamically regulated during glioblastoma multiforme tumorigenesis

Authors: Maria Cecília Oliveira-Nunes, Suzana Assad Kahn, Ana Luiza de Oliveira Barbeitas, Tania Cristina Leite de Sampaio e Spohr, Luiz Gustavo Feijó Dubois, Grasiella Maria Ventura Matioszek, William Querido, Loraine Campanati, José Marques de Brito Neto, Flavia Regina Souza Lima, Vivaldo Moura-Neto, Katia Carneiro

Published in: Cancer Cell International | Issue 1/2016

Login to get access

Abstract

Background

Glioblastoma (GBM) is the most common primary brain tumor presenting self-renewing cancer stem cells. The role of these cells on the development of the tumors has been proposed to recapitulate programs from embryogenesis. Recently, the embryonic transforming growth factor-β (TGF-β) protein Nodal has been shown to be reactivated upon tumor development; however, its availability in GBM cells has not been addressed so far. In this study, we investigated by an original approach the mechanisms that dynamically control both intra and extracellular Nodal availability during GBM tumorigenesis.

Methods

We characterized the dynamics of Nodal availability in both stem and more differentiated GBM cells through morphological analysis, immunofluorescence of Nodal protein and of early (EEA1 and Rab5) and late (Rab7 and Rab11) endocytic markers and Western Blot. Tukey’s test was used to analyze the prevalent correlation of Nodal with different endocytic markers inside specific differentiation states, and Sidak’s multiple comparisons test was used to compare the prevalence of Nodal/endocytic markers co-localization between two differentiation states of GBM cells. Paired t test was used to analyze the abundance of Nodal protein, in extra and intracellular media.

Results

The cytoplasmic distribution of Nodal was dynamically regulated and strongly correlated with the differentiation status of GBM cells. While Nodal-positive vesicle-like particles were symmetrically distributed in GBM stem cells (GBMsc), they presented asymmetric perinuclear localization in more differentiated GBM cells (mdGBM). Strikingly, when subjected to dedifferentiation, the distribution of Nodal in mdGBM shifted to a symmetric pattern. Moreover, the availability of both intracellular and secreted Nodal were downregulated upon GBMsc differentiation, with cells becoming elongated, negative for Nodal and positive for Nestin. Interestingly, the co-localization of Nodal with endosomal vesicles also depended on the differentiation status of the cells, with Nodal seen more packed in EEA1/Rab5 + vesicles in GBMsc and more in Rab7/11 + vesicles in mdGBM.

Conclusions

Our results show for the first time that Nodal availability relates to GBM cell differentiation status and that it is dynamically regulated by an endocytic pathway during GBM tumorigenesis, shedding new light on molecular pathways that might emerge as putative targets for Nodal signaling in GBM therapy.
Appendix
Available only for authorised users
Literature
1.
go back to reference Levin M, Johnson RL, Stern CD, Kuehn M, Tabin C. A molecular pathway determining left-right asymmetry in chick embryogenesis. Cell. 1995;82:803–14.CrossRefPubMed Levin M, Johnson RL, Stern CD, Kuehn M, Tabin C. A molecular pathway determining left-right asymmetry in chick embryogenesis. Cell. 1995;82:803–14.CrossRefPubMed
2.
go back to reference Levin M, Pagan S, Roberts DJ, Cooke J, Kuehn MR, Tabin CJ. Left/right patterning signals and the independent regulation of different aspects of situs in the chick embryo. Dev Biol. 1997;189:57–67.CrossRefPubMed Levin M, Pagan S, Roberts DJ, Cooke J, Kuehn MR, Tabin CJ. Left/right patterning signals and the independent regulation of different aspects of situs in the chick embryo. Dev Biol. 1997;189:57–67.CrossRefPubMed
3.
go back to reference Carneiro K, Donnet C, Rejtar T, Karger BL, Barisone GA, Diaz E, Kortagere S, Lemire JM, Levin M. Histone deacetylase activity is necessary for left-right patterning during vertebrate development. BMC Dev Biol. 2011;11:29.CrossRefPubMedPubMedCentral Carneiro K, Donnet C, Rejtar T, Karger BL, Barisone GA, Diaz E, Kortagere S, Lemire JM, Levin M. Histone deacetylase activity is necessary for left-right patterning during vertebrate development. BMC Dev Biol. 2011;11:29.CrossRefPubMedPubMedCentral
4.
go back to reference Galvin-Burgess KE, Travis ED, Pierson KE, Vivian JL. TGF-beta-superfamily signaling regulates embryonic stem cell heterogeneity: self-renewal as a dynamic and regulated equilibrium. Stem Cells. 2013;31:48–58.CrossRefPubMedPubMedCentral Galvin-Burgess KE, Travis ED, Pierson KE, Vivian JL. TGF-beta-superfamily signaling regulates embryonic stem cell heterogeneity: self-renewal as a dynamic and regulated equilibrium. Stem Cells. 2013;31:48–58.CrossRefPubMedPubMedCentral
5.
go back to reference James D, Levine AJ, Besser D, Hemmati-Brivanlou A. TGFbeta/activin/nodal signaling is necessary for the maintenance of pluripotency in human embryonic stem cells. Development. 2005;132:1273–82.CrossRefPubMed James D, Levine AJ, Besser D, Hemmati-Brivanlou A. TGFbeta/activin/nodal signaling is necessary for the maintenance of pluripotency in human embryonic stem cells. Development. 2005;132:1273–82.CrossRefPubMed
6.
go back to reference Strizzi L, Hardy KM, Margaryan NV, Hillman DW, Seftor EA, Chen B, Geiger XJ, Thompson EA, Lingle WL, Andorfer CA, Perez EA, Hendrix MJ. Potential for the embryonic morphogen Nodal as a prognostic and predictive biomarker in breast cancer. Breast Cancer Res. 2012;14:R75.CrossRefPubMedPubMedCentral Strizzi L, Hardy KM, Margaryan NV, Hillman DW, Seftor EA, Chen B, Geiger XJ, Thompson EA, Lingle WL, Andorfer CA, Perez EA, Hendrix MJ. Potential for the embryonic morphogen Nodal as a prognostic and predictive biomarker in breast cancer. Breast Cancer Res. 2012;14:R75.CrossRefPubMedPubMedCentral
7.
go back to reference Vo BT, Khan SA. Expression of nodal and nodal receptors in prostate stem cells and prostate cancer cells: autocrine effects on cell proliferation and migration. Prostate. 2011;71:1084–96.CrossRefPubMedPubMedCentral Vo BT, Khan SA. Expression of nodal and nodal receptors in prostate stem cells and prostate cancer cells: autocrine effects on cell proliferation and migration. Prostate. 2011;71:1084–96.CrossRefPubMedPubMedCentral
8.
go back to reference Hueng DY, Lin GJ, Huang SH, Liu LW, Ju DT, Chen YW, Sytwu HK, Chang C, Huang SM, Yeh YS, Lee HM, Ma HI. Inhibition of Nodal suppresses angiogenesis and growth of human gliomas. J Neurooncol. 2011;104:21–31.CrossRefPubMed Hueng DY, Lin GJ, Huang SH, Liu LW, Ju DT, Chen YW, Sytwu HK, Chang C, Huang SM, Yeh YS, Lee HM, Ma HI. Inhibition of Nodal suppresses angiogenesis and growth of human gliomas. J Neurooncol. 2011;104:21–31.CrossRefPubMed
9.
go back to reference McAllister JC, Zhan Q, Weishaupt C, Hsu MY, Murphy GF. The embryonic morphogen, nodal, is associated with channel-like structures in human malignant melanoma xenografts. J Cutan Pathol. 2010;37(Suppl 1):19–25.CrossRefPubMedPubMedCentral McAllister JC, Zhan Q, Weishaupt C, Hsu MY, Murphy GF. The embryonic morphogen, nodal, is associated with channel-like structures in human malignant melanoma xenografts. J Cutan Pathol. 2010;37(Suppl 1):19–25.CrossRefPubMedPubMedCentral
10.
go back to reference Strizzi L, Hardy KM, Kirsammer GT, Gerami P, Hendrix MJ. Embryonic signaling in melanoma: potential for diagnosis and therapy. Lab Invest. 2011;91:819–24.CrossRefPubMedPubMedCentral Strizzi L, Hardy KM, Kirsammer GT, Gerami P, Hendrix MJ. Embryonic signaling in melanoma: potential for diagnosis and therapy. Lab Invest. 2011;91:819–24.CrossRefPubMedPubMedCentral
11.
go back to reference Topczewska JM, Postovit LM, Margaryan NV, Sam A, Hess AR, Wheaton WW, Nickoloff BJ, Topczewski J, Hendrix MJ. Embryonic and tumorigenic pathways converge via Nodal signaling: role in melanoma aggressiveness. Nat Med. 2006;12:925–32.CrossRefPubMed Topczewska JM, Postovit LM, Margaryan NV, Sam A, Hess AR, Wheaton WW, Nickoloff BJ, Topczewski J, Hendrix MJ. Embryonic and tumorigenic pathways converge via Nodal signaling: role in melanoma aggressiveness. Nat Med. 2006;12:925–32.CrossRefPubMed
12.
go back to reference Xu G, Zhong Y, Munir S, Yang BB, Tsang BK, Peng C. Nodal induces apoptosis and inhibits proliferation in human epithelial ovarian cancer cells via activin receptor-like kinase 7. J Clin Endocrinol Metab. 2004;89:5523–34.CrossRefPubMed Xu G, Zhong Y, Munir S, Yang BB, Tsang BK, Peng C. Nodal induces apoptosis and inhibits proliferation in human epithelial ovarian cancer cells via activin receptor-like kinase 7. J Clin Endocrinol Metab. 2004;89:5523–34.CrossRefPubMed
13.
go back to reference Xu G, Zhou H, Wang Q, Auersperg N, Peng C. Activin receptor-like kinase 7 induces apoptosis through up-regulation of Bax and down-regulation of Xiap in normal and malignant ovarian epithelial cell lines. Mol Cancer Res. 2006;4:235–46.CrossRefPubMed Xu G, Zhou H, Wang Q, Auersperg N, Peng C. Activin receptor-like kinase 7 induces apoptosis through up-regulation of Bax and down-regulation of Xiap in normal and malignant ovarian epithelial cell lines. Mol Cancer Res. 2006;4:235–46.CrossRefPubMed
14.
go back to reference Ye G, Fu G, Cui S, Zhao S, Bernaudo S, Bai Y, Ding Y, Zhang Y, Yang BB, Peng C. MicroRNA 376c enhances ovarian cancer cell survival by targeting activin receptor-like kinase 7: implications for chemoresistance. J Cell Sci. 2011;124:359–68.CrossRefPubMed Ye G, Fu G, Cui S, Zhao S, Bernaudo S, Bai Y, Ding Y, Zhang Y, Yang BB, Peng C. MicroRNA 376c enhances ovarian cancer cell survival by targeting activin receptor-like kinase 7: implications for chemoresistance. J Cell Sci. 2011;124:359–68.CrossRefPubMed
15.
go back to reference Zhong Y, Xu G, Ye G, Lee D, Modica-Amore J, Peng C. Nodal and activin receptor-like kinase 7 induce apoptosis in human breast cancer cell lines: role of caspase 3. Int J Physiol Pathophysiol Pharmacol. 2009;1:83–96.PubMedPubMedCentral Zhong Y, Xu G, Ye G, Lee D, Modica-Amore J, Peng C. Nodal and activin receptor-like kinase 7 induce apoptosis in human breast cancer cell lines: role of caspase 3. Int J Physiol Pathophysiol Pharmacol. 2009;1:83–96.PubMedPubMedCentral
16.
go back to reference Alves TR, Lima FR, Kahn SA, Lobo D, Dubois LG, Soletti R, Borges H, Neto VM. Glioblastoma cells: a heterogeneous and fatal tumor interacting with the parenchyma. Life Sci. 2011;89:532–9.CrossRefPubMed Alves TR, Lima FR, Kahn SA, Lobo D, Dubois LG, Soletti R, Borges H, Neto VM. Glioblastoma cells: a heterogeneous and fatal tumor interacting with the parenchyma. Life Sci. 2011;89:532–9.CrossRefPubMed
17.
go back to reference Lima FR, Kahn SA, Soletti RC, Biasoli D, Alves T, da Fonseca AC, Garcia C, Romao L, Brito J, Holanda-Afonso R, Faria J, Borges H, Moura-Neto V. Glioblastoma: therapeutic challenges, what lies ahead. Biochim Biophys Acta. 1826;2012:338–49. Lima FR, Kahn SA, Soletti RC, Biasoli D, Alves T, da Fonseca AC, Garcia C, Romao L, Brito J, Holanda-Afonso R, Faria J, Borges H, Moura-Neto V. Glioblastoma: therapeutic challenges, what lies ahead. Biochim Biophys Acta. 1826;2012:338–49.
18.
go back to reference Maher EA, Furnari FB, Bachoo RM, Rowitch DH, Louis DN, Cavenee WK, DePinho RA. Malignant glioma: genetics and biology of a grave matter. Genes Dev. 2001;15:1311–33.CrossRefPubMed Maher EA, Furnari FB, Bachoo RM, Rowitch DH, Louis DN, Cavenee WK, DePinho RA. Malignant glioma: genetics and biology of a grave matter. Genes Dev. 2001;15:1311–33.CrossRefPubMed
20.
go back to reference Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, Vescovi A. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res. 2004;64:7011–21.CrossRefPubMed Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, Vescovi A. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res. 2004;64:7011–21.CrossRefPubMed
21.
go back to reference Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature. 2004;432:396–401.CrossRefPubMed Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature. 2004;432:396–401.CrossRefPubMed
22.
go back to reference Persano L, Rampazzo E, Basso G, Viola G. Glioblastoma cancer stem cells: role of the microenvironment and therapeutic targeting. Biochem Pharmacol. 2013;85:612–22.CrossRefPubMed Persano L, Rampazzo E, Basso G, Viola G. Glioblastoma cancer stem cells: role of the microenvironment and therapeutic targeting. Biochem Pharmacol. 2013;85:612–22.CrossRefPubMed
23.
go back to reference Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444:756–60.CrossRefPubMed Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444:756–60.CrossRefPubMed
24.
go back to reference Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, Qian D, Lam JS, Ailles LE, Wong M, Joshua B, Kaplan MJ, Wapnir I, Dirbas FM, Somlo G, Garberoglio C, Paz B, Shen J, Lau SK, Quake SR, Brown JM, Weissman IL, Clarke MF. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 2009;458:780–3.CrossRefPubMedPubMedCentral Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, Qian D, Lam JS, Ailles LE, Wong M, Joshua B, Kaplan MJ, Wapnir I, Dirbas FM, Somlo G, Garberoglio C, Paz B, Shen J, Lau SK, Quake SR, Brown JM, Weissman IL, Clarke MF. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 2009;458:780–3.CrossRefPubMedPubMedCentral
25.
26.
go back to reference Kahn SA, Biasoli D, Garcia C, Geraldo LH, Pontes B, Sobrinho M, Frauches AC, Romao L, Soletti RC, Assuncao Fdos S, Tovar-Moll F, de Souza JM, Lima FR, Anderluh G, Moura-neto V. Equinatoxin II potentiates temozolomide- and etoposide-induced glioblastoma cell death. Curr Top Med Chem. 2012;12:2082–93.CrossRefPubMed Kahn SA, Biasoli D, Garcia C, Geraldo LH, Pontes B, Sobrinho M, Frauches AC, Romao L, Soletti RC, Assuncao Fdos S, Tovar-Moll F, de Souza JM, Lima FR, Anderluh G, Moura-neto V. Equinatoxin II potentiates temozolomide- and etoposide-induced glioblastoma cell death. Curr Top Med Chem. 2012;12:2082–93.CrossRefPubMed
27.
go back to reference Faria J, Romao L, Martins S, Alves T, Mendes FA, de Faria GP, Hollanda R, Takiya C, Chimelli L, Morandi V, de Souza JM, Abreu JG. Moura Neto V. Interactive properties of human glioblastoma cells with brain neurons in culture and neuronal modulation of glial laminin organization. Differentiation. 2006;74:562–72.CrossRefPubMed Faria J, Romao L, Martins S, Alves T, Mendes FA, de Faria GP, Hollanda R, Takiya C, Chimelli L, Morandi V, de Souza JM, Abreu JG. Moura Neto V. Interactive properties of human glioblastoma cells with brain neurons in culture and neuronal modulation of glial laminin organization. Differentiation. 2006;74:562–72.CrossRefPubMed
28.
go back to reference Biasoli D, Kahn SA, Cornelio TA, Furtado M, Campanati L, Chneiweiss H, Moura-Neto V, Borges HL. Retinoblastoma protein regulates the crosstalk between autophagy and apoptosis, and favors glioblastoma resistance to etoposide. Cell Death Dis. 2013;4:e767.CrossRefPubMedPubMedCentral Biasoli D, Kahn SA, Cornelio TA, Furtado M, Campanati L, Chneiweiss H, Moura-Neto V, Borges HL. Retinoblastoma protein regulates the crosstalk between autophagy and apoptosis, and favors glioblastoma resistance to etoposide. Cell Death Dis. 2013;4:e767.CrossRefPubMedPubMedCentral
29.
go back to reference Chinnaiyan P, Wang M, Rojiani AM, Tofilon PJ, Chakravarti A, Ang KK, Zhang HZ, Hammond E, Curran W Jr, Mehta MP. The prognostic value of nestin expression in newly diagnosed glioblastoma: report from the Radiation Therapy Oncology Group. Radiat Oncol. 2008;3:32.CrossRefPubMedPubMedCentral Chinnaiyan P, Wang M, Rojiani AM, Tofilon PJ, Chakravarti A, Ang KK, Zhang HZ, Hammond E, Curran W Jr, Mehta MP. The prognostic value of nestin expression in newly diagnosed glioblastoma: report from the Radiation Therapy Oncology Group. Radiat Oncol. 2008;3:32.CrossRefPubMedPubMedCentral
30.
go back to reference Blanchet MH, Le Good JA, Oorschot V, Baflast S, Minchiotti G, Klumperman J, Constam DB. Cripto localizes Nodal at the limiting membrane of early endosomes. Sci Signal. 2008;1:ra13.CrossRefPubMed Blanchet MH, Le Good JA, Oorschot V, Baflast S, Minchiotti G, Klumperman J, Constam DB. Cripto localizes Nodal at the limiting membrane of early endosomes. Sci Signal. 2008;1:ra13.CrossRefPubMed
31.
go back to reference Ng EL, Gan BQ, Ng F, Tang BL. Rab GTPases regulating receptor trafficking at the late endosome-lysosome membranes. Cell Biochem Funct. 2012;30:515–23.CrossRefPubMed Ng EL, Gan BQ, Ng F, Tang BL. Rab GTPases regulating receptor trafficking at the late endosome-lysosome membranes. Cell Biochem Funct. 2012;30:515–23.CrossRefPubMed
33.
go back to reference Sandri C, Caccavari F, Valdembri D, Camillo C, Veltel S, Santambrogio M, Lanzetti L, Bussolino F, Ivaska J, Serini G. The R-Ras/RIN2/Rab5 complex controls endothelial cell adhesion and morphogenesis via active integrin endocytosis and Rac signaling. Cell Res. 2012;22:1479–501.CrossRefPubMedPubMedCentral Sandri C, Caccavari F, Valdembri D, Camillo C, Veltel S, Santambrogio M, Lanzetti L, Bussolino F, Ivaska J, Serini G. The R-Ras/RIN2/Rab5 complex controls endothelial cell adhesion and morphogenesis via active integrin endocytosis and Rac signaling. Cell Res. 2012;22:1479–501.CrossRefPubMedPubMedCentral
34.
go back to reference Frittoli E, Palamidessi A, Marighetti P, Confalonieri S, Bianchi F, Malinverno C, Mazzarol G, Viale G, Martin-Padura I, Garre M, Parazzoli D, Mattei V, Cortellino S, Bertalot G, Di Fiore PP, Scita G. A RAB5/RAB4 recycling circuitry induces a proteolytic invasive program and promotes tumor dissemination. J Cell Biol. 2014;206:307–28.CrossRefPubMedPubMedCentral Frittoli E, Palamidessi A, Marighetti P, Confalonieri S, Bianchi F, Malinverno C, Mazzarol G, Viale G, Martin-Padura I, Garre M, Parazzoli D, Mattei V, Cortellino S, Bertalot G, Di Fiore PP, Scita G. A RAB5/RAB4 recycling circuitry induces a proteolytic invasive program and promotes tumor dissemination. J Cell Biol. 2014;206:307–28.CrossRefPubMedPubMedCentral
37.
38.
go back to reference Zhang M, Chen L, Wang S, Wang T. Rab7: roles in membrane trafficking and disease. Biosci Rep. 2009;29:193–209.CrossRefPubMed Zhang M, Chen L, Wang S, Wang T. Rab7: roles in membrane trafficking and disease. Biosci Rep. 2009;29:193–209.CrossRefPubMed
39.
go back to reference Vallier L, Reynolds D, Pedersen RA. Nodal inhibits differentiation of human embryonic stem cells along the neuroectodermal default pathway. Dev Biol. 2004;275:403–21.CrossRefPubMed Vallier L, Reynolds D, Pedersen RA. Nodal inhibits differentiation of human embryonic stem cells along the neuroectodermal default pathway. Dev Biol. 2004;275:403–21.CrossRefPubMed
Metadata
Title
The availability of the embryonic TGF-β protein Nodal is dynamically regulated during glioblastoma multiforme tumorigenesis
Authors
Maria Cecília Oliveira-Nunes
Suzana Assad Kahn
Ana Luiza de Oliveira Barbeitas
Tania Cristina Leite de Sampaio e Spohr
Luiz Gustavo Feijó Dubois
Grasiella Maria Ventura Matioszek
William Querido
Loraine Campanati
José Marques de Brito Neto
Flavia Regina Souza Lima
Vivaldo Moura-Neto
Katia Carneiro
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2016
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-016-0324-3

Other articles of this Issue 1/2016

Cancer Cell International 1/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine